1
|
Jamerson LE, Bradshaw PC. The Roles of White Adipose Tissue and Liver NADPH in Dietary Restriction-Induced Longevity. Antioxidants (Basel) 2024; 13:820. [PMID: 39061889 PMCID: PMC11273496 DOI: 10.3390/antiox13070820] [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: 05/29/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Dietary restriction (DR) protocols frequently employ intermittent fasting. Following a period of fasting, meal consumption increases lipogenic gene expression, including that of NADPH-generating enzymes that fuel lipogenesis in white adipose tissue (WAT) through the induction of transcriptional regulators SREBP-1c and CHREBP. SREBP-1c knockout mice, unlike controls, did not show an extended lifespan on the DR diet. WAT cytoplasmic NADPH is generated by both malic enzyme 1 (ME1) and the pentose phosphate pathway (PPP), while liver cytoplasmic NADPH is primarily synthesized by folate cycle enzymes provided one-carbon units through serine catabolism. During the daily fasting period of the DR diet, fatty acids are released from WAT and are transported to peripheral tissues, where they are used for beta-oxidation and for phospholipid and lipid droplet synthesis, where monounsaturated fatty acids (MUFAs) may activate Nrf1 and inhibit ferroptosis to promote longevity. Decreased WAT NADPH from PPP gene knockout stimulated the browning of WAT and protected from a high-fat diet, while high levels of NADPH-generating enzymes in WAT and macrophages are linked to obesity. But oscillations in WAT [NADPH]/[NADP+] from feeding and fasting cycles may play an important role in maintaining metabolic plasticity to drive longevity. Studies measuring the WAT malate/pyruvate as a proxy for the cytoplasmic [NADPH]/[NADP+], as well as studies using fluorescent biosensors expressed in the WAT of animal models to monitor the changes in cytoplasmic [NADPH]/[NADP+], are needed during ad libitum and DR diets to determine the changes that are associated with longevity.
Collapse
Affiliation(s)
| | - Patrick C. Bradshaw
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| |
Collapse
|
2
|
Chacón CF, Parachú Marcó MV, Poletta GL, Siroski PA. Lipid metabolism in crocodilians: A field with promising applications in the field of ecotoxicology. ENVIRONMENTAL RESEARCH 2024; 252:119017. [PMID: 38704009 DOI: 10.1016/j.envres.2024.119017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/06/2024]
Abstract
In the last years, lipid physiology has become an important research target for systems biology applied to the field of ecotoxicology. Lipids are not only essential components of biological membranes, but also participate in extra and intracellular signaling processes and as signal transducers and amplifiers of regulatory cascades. Particularly in sauropsids, lipids are the main source of energy for reproduction, growth, and embryonic development. In nature, organisms are exposed to different stressors, such as parasites, diseases and environmental contaminants, which interact with lipid signaling and metabolic pathways, disrupting lipid homeostasis. The system biology approach applied to ecotoxicological studies is crucial to evaluate metabolic regulation under environmental stress produced by xenobiotics. In this review, we cover information of molecular mechanisms that contribute to lipid metabolism homeostasis in sauropsids, specifically in crocodilian species. We focus on the role of lipid metabolism as a powerful source of energy and its importance during oocyte maturation, which has been increasingly recognized in many species, but information is still scarce in crocodiles. Finally, we highlight priorities for future research on the influence of environmental stressors on lipid metabolism, their potential effect on the reproductive system and thus on the offspring, and their implications on crocodilians conservation.
Collapse
Affiliation(s)
- C F Chacón
- Laboratorio de Ecología Molecular Aplicada (LEMA), Instituto de Ciencias Veterinarias del Litoral- Consejo Nacional de Investigaciones Científicas y Técnicas (ICiVet Litoral-CONICET/UNL), Av. Aristóbulo del Valle 8700, 3000, Santa Fe, Argentina; Proyecto Yacaré (MAyCC, Gob. de Santa Fe), Av. Aristóbulo del Valle 8700, 3000, Santa Fe, Argentina.
| | - M V Parachú Marcó
- Laboratorio de Ecología Molecular Aplicada (LEMA), Instituto de Ciencias Veterinarias del Litoral- Consejo Nacional de Investigaciones Científicas y Técnicas (ICiVet Litoral-CONICET/UNL), Av. Aristóbulo del Valle 8700, 3000, Santa Fe, Argentina; Proyecto Yacaré (MAyCC, Gob. de Santa Fe), Av. Aristóbulo del Valle 8700, 3000, Santa Fe, Argentina
| | - G L Poletta
- Laboratorio de Ecología Molecular Aplicada (LEMA), Instituto de Ciencias Veterinarias del Litoral- Consejo Nacional de Investigaciones Científicas y Técnicas (ICiVet Litoral-CONICET/UNL), Av. Aristóbulo del Valle 8700, 3000, Santa Fe, Argentina; Toxicología, Farmacología y Bioquímica Legal, FBCB-UNL, CONICET, Ciudad Universitaria, Paraje El Pozo S/N, 3000, Santa Fe, Argentina
| | - P A Siroski
- Laboratorio de Ecología Molecular Aplicada (LEMA), Instituto de Ciencias Veterinarias del Litoral- Consejo Nacional de Investigaciones Científicas y Técnicas (ICiVet Litoral-CONICET/UNL), Av. Aristóbulo del Valle 8700, 3000, Santa Fe, Argentina; Proyecto Yacaré (MAyCC, Gob. de Santa Fe), Av. Aristóbulo del Valle 8700, 3000, Santa Fe, Argentina
| |
Collapse
|
3
|
Matsumura S, Signoretti C, Fatehi S, Tumenbayar BI, D'Addario C, Nimmer E, Thomas C, Viswanathan T, Wolf A, Garcia V, Rocic P, Bae Y, Alam SS, Gupte SA. Loss-of-function G6PD variant moderated high-fat diet-induced obesity, adipocyte hypertrophy, and fatty liver in male rats. J Biol Chem 2024; 300:107460. [PMID: 38876306 PMCID: PMC11328872 DOI: 10.1016/j.jbc.2024.107460] [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: 10/04/2023] [Revised: 05/11/2024] [Accepted: 05/21/2024] [Indexed: 06/16/2024] Open
Abstract
Obesity is a major risk factor for liver and cardiovascular diseases. However, obesity-driven mechanisms that contribute to the pathogenesis of multiple organ diseases are still obscure and treatment is inadequate. We hypothesized that increased , glucose-6-phosphate dehydrogenase (G6PD), the key rate-limiting enzyme in the pentose shunt, is critical in evoking metabolic reprogramming in multiple organs and is a significant contributor to the pathogenesis of liver and cardiovascular diseases. G6PD is induced by a carbohydrate-rich diet and insulin. Long-term (8 months) high-fat diet (HFD) feeding increased body weight and elicited metabolic reprogramming in visceral fat, liver, and aorta, of the wild-type rats. In addition, HFD increased inflammatory chemokines in visceral fat. Interestingly, CRISPR-edited loss-of-function Mediterranean G6PD variant (G6PDS188F) rats, which mimic human polymorphism, moderated HFD-induced weight gain and metabolic reprogramming in visceral fat, liver, and aorta. The G6PDS188F variant prevented HFD-induced CCL7 and adipocyte hypertrophy. Furthermore, the G6PDS188F variant increased Magel2 - a gene encoding circadian clock-related protein that suppresses obesity associated with Prader-Willi syndrome - and reduced HFD-induced non-alcoholic fatty liver. Additionally, the G6PDS188F variant reduced aging-induced aortic stiffening. Our findings suggest G6PD is a regulator of HFD-induced obesity, adipocyte hypertrophy, and fatty liver.
Collapse
Affiliation(s)
- Shun Matsumura
- Department of Pharmacology, New York Medical College, Valhalla, New York, USA
| | | | - Samuel Fatehi
- Department of Pharmacology, New York Medical College, Valhalla, New York, USA
| | - Bat Ider Tumenbayar
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Catherine D'Addario
- Department of Pharmacology, New York Medical College, Valhalla, New York, USA
| | - Erik Nimmer
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Colin Thomas
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Trisha Viswanathan
- Department of Pharmacology, New York Medical College, Valhalla, New York, USA
| | - Alexandra Wolf
- Department of Pharmacology, New York Medical College, Valhalla, New York, USA
| | - Victor Garcia
- Department of Pharmacology, New York Medical College, Valhalla, New York, USA
| | - Petra Rocic
- Department of Physiology & Pharmacology, SHSU College of Osteopathic Medicine, Conroe, Texas, USA
| | - Yongho Bae
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA; Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Sm Shafiqul Alam
- Department of Pathology, Microbiology, and Immunology (PMI), New York Medical College, Valhalla, New York, USA
| | - Sachin A Gupte
- Department of Pharmacology, New York Medical College, Valhalla, New York, USA.
| |
Collapse
|
4
|
Wupperfeld D, Fricker G, Bois De Fer B, Popovic B. Essential phospholipids impact cytokine secretion and alter lipid-metabolizing enzymes in human hepatocyte cell lines. Pharmacol Rep 2024; 76:572-584. [PMID: 38664334 PMCID: PMC11126482 DOI: 10.1007/s43440-024-00595-4] [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: 11/23/2023] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 05/09/2024]
Abstract
BACKGROUND Essential phospholipids (EPL) are hepatoprotective. METHODS The effects on interleukin (IL)-6 and -8 secretion and on certain lipid-metabolizing enzymes of non-cytotoxic concentrations of EPL (0.1 and 0.25 mg/ml), polyenylphosphatidylcholine (PPC), and phosphatidylinositol (PtdIns) (both at 0.1 and 1 mg/ml), compared with untreated controls, were assessed in human hepatocyte cell lines (HepG2, HepaRG, and steatotic HepaRG). RESULTS Lipopolysaccharide (LPS)-induced IL-6 secretion was significantly decreased in HepaRG cells by most phospholipids, and significantly increased in steatotic HepaRG cells with at least one concentration of EPL and PtdIns. LPS-induced IL-8 secretion was significantly increased in HepaRG and steatotic HepaRG cells with all phospholipids. All phospholipids significantly decreased amounts of fatty acid synthase in steatotic HepaRG cells and the amounts of acyl-CoA oxidase in HepaRG cells. Amounts of lecithin cholesterol acyltransferase were significantly decreased in HepG2 and HepaRG cells by most phospholipids, and significantly increased with 0.1 mg/ml PPC (HepaRG cells) and 1 mg/ml PtdIns (steatotic HepaRG cells). Glucose-6-phosphate dehydrogenase activity was unaffected by any phospholipid in any cell line. CONCLUSIONS EPL, PPC, and PtdIns impacted the secretion of pro-inflammatory cytokines and affected amounts of several key lipid-metabolizing enzymes in human hepatocyte cell lines. Such changes may help liver function improvement, and provide further insights into the EPL's mechanism of action.
Collapse
Affiliation(s)
- Dominik Wupperfeld
- Department of Pharmaceutical Technology and Biopharmacy, Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls University of Heidelberg, Heidelberg, Germany
| | - Gert Fricker
- Department of Pharmaceutical Technology and Biopharmacy, Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls University of Heidelberg, Heidelberg, Germany
| | | | - Branko Popovic
- Sanofi, Frankfurt am Main, K607, 65929, Industriepark Hoechst, Germany.
| |
Collapse
|
5
|
Soskic MB, Zakic T, Korac A, Korac B, Jankovic A. Metabolic remodeling of visceral and subcutaneous white adipose tissue during reacclimation of rats after cold. Appl Physiol Nutr Metab 2024; 49:649-658. [PMID: 38241659 DOI: 10.1139/apnm-2023-0448] [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] [Indexed: 01/21/2024]
Abstract
Deciphering lipid metabolism in white adipose tissue (WAT) depots during weight gain is important to understand the heterogeneity of WAT and its roles in obesity. Here, we examined the expression of key enzymes of lipid metabolism and changes in the morphology of representative visceral (epididymal) and subcutaneous (inguinal) WAT (eWAT and iWAT, respectively)-in adult male rats acclimated to cold (4 ± 1 °C) for 45 days and reacclimated to room temperature (RT, 22 ± 1 °C) for 1, 3, 7, 12, 21, or 45 days. The relative mass of both depots decreased to a similar extent after cold acclimation. However, fatty acid synthase (FAS), glucose-6-phosphate dehydrogenase (G6PDH), and medium-chain acyl-CoA dehydrogenase (ACADM) protein level increased only in eWAT, whereas adipose triglyceride lipase (ATGL) expression increased only in iWAT. During reacclimation, the relative mass of eWAT reached control values on day 12 and that of iWAT on day 45 of reacclimation. The faster recovery of eWAT mass is associated with higher expression of FAS, acetyl-CoA carboxylase (ACC), G6PDH, and ACADM during reacclimation and a delayed increase in ATGL. The absence of an increase in proliferating cell nuclear antigen suggests that the observed depot-specific mass increase is predominantly due to metabolic adjustments. In summary, this study shows a differential rate of visceral and subcutaneous adipose tissue weight regain during post-cold reacclimation of rats at RT. Faster recovery of the visceral WAT as compared to subcutaneous WAT during reacclimation at RT could be attributed to observed differences in the expression patterns of lipid metabolic enzymes.
Collapse
Affiliation(s)
- Marta Budnar Soskic
- Department of Physiology, Institute for Biological Research "Sinisa Stankovic"-National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| | - Tamara Zakic
- Department of Physiology, Institute for Biological Research "Sinisa Stankovic"-National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| | - Aleksandra Korac
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia
| | - Bato Korac
- Department of Physiology, Institute for Biological Research "Sinisa Stankovic"-National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia
| | - Aleksandra Jankovic
- Department of Physiology, Institute for Biological Research "Sinisa Stankovic"-National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
6
|
Chen G, Lin Z, Peng H, Zhang S, Zhang Z, Zhang X, Nie Q, Luo W. The transmembrane protein TMEM182 promotes fat deposition and alters metabolomics and lipidomics. Int J Biol Macromol 2024; 259:129144. [PMID: 38181918 DOI: 10.1016/j.ijbiomac.2023.129144] [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: 10/29/2023] [Revised: 12/10/2023] [Accepted: 12/28/2023] [Indexed: 01/07/2024]
Abstract
TMEM182, a transmembrane protein highly expressed in muscle and adipose tissues, plays a crucial role in muscle cell differentiation, metabolism, and signaling. However, its role in fat deposition and metabolism is still unknown. In this study, we used overexpression and knockout models to examine the impact of TMEM182 on fat synthesis and metabolism. Our results showed that TMEM182 overexpression increased the expression of fat synthesis-related genes and promoted the differentiation of preadipocytes into fat cells. In TMEM182 knockout mice, there was a significant decrease in abdominal fat deposition. RNA sequencing results showed that TMEM182 overexpression in preadipocytes enhanced the activity of pathways related to fat formation, ECM-receptor interaction, and cell adhesion. Furthermore, our analysis using UPLC-MS/MS showed that TMEM182 significantly altered the metabolite and lipid content and composition in chicken breast muscle. Specifically, TMEM182 increased the content of amino acids and their derivatives in chicken breast muscle, promoting amino acid metabolic pathways. Lipidomics also revealed a significant increase in the content of glycerophospholipids, sphingolipids, and phospholipids in the breast muscle after TMEM182 overexpression. These findings suggest that TMEM182 plays a crucial role in regulating fat deposition and metabolism, making it a potential target for treating obesity-related diseases and animal breeding.
Collapse
Affiliation(s)
- Genghua Chen
- College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China; State Key Laboratory of Livestock and Poultry Breeding, Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Zetong Lin
- College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China; State Key Laboratory of Livestock and Poultry Breeding, Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Haoqi Peng
- College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China; State Key Laboratory of Livestock and Poultry Breeding, Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Shuai Zhang
- College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China; State Key Laboratory of Livestock and Poultry Breeding, Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Zihao Zhang
- College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China; State Key Laboratory of Livestock and Poultry Breeding, Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Xiquan Zhang
- College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China; State Key Laboratory of Livestock and Poultry Breeding, Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Qinghua Nie
- College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China; State Key Laboratory of Livestock and Poultry Breeding, Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Wen Luo
- College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China; State Key Laboratory of Livestock and Poultry Breeding, Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
7
|
Lamichhane G, Liu J, Lee SJ, Lee DY, Zhang G, Kim Y. Curcumin Mitigates the High-Fat High-Sugar Diet-Induced Impairment of Spatial Memory, Hepatic Metabolism, and the Alteration of the Gut Microbiome in Alzheimer's Disease-Induced (3xTg-AD) Mice. Nutrients 2024; 16:240. [PMID: 38257133 PMCID: PMC10818691 DOI: 10.3390/nu16020240] [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: 12/21/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
The escalating prevalence of metabolic diseases and an aging demographic has been correlated with a concerning rise in Alzheimer's disease (AD) incidence. This study aimed to access the protective effects of curcumin, a bioactive flavonoid from turmeric, on spatial memory, metabolic functions, and the regulation of the gut microbiome in AD-induced (3xTg-AD) mice fed with either a normal chow diet (NCD) or a high-fat high-sugar diet (HFHSD). Our findings revealed an augmented susceptibility of the HFHSD-fed 3xTg-AD mice for weight gain and memory impairment, while curcumin supplementation demonstrated a protective effect against these changes. This was evidenced by significantly reduced body weight gain and improved behavioral and cognitive function in the curcumin-treated group. These improvements were substantiated by diminished fatty acid synthesis, altered cholesterol metabolism, and suppressed adipogenesis-related pathways in the liver, along with modified synaptic plasticity-related pathways in the brain. Moreover, curcumin enriched beneficial gut microbiota, including Oscillospiraceae and Rikenellaceae at the family level, and Oscillibacter, Alistipes, Pseudoflavonifractor, Duncaniella, and Flintibacter at the genus level. The observed alteration in these gut microbiota profiles suggests a potential crosswalk in the liver and brain for regulating metabolic and cognitive functions, particularly in the context of obesity-associated cognitive disfunction, notably AD.
Collapse
Affiliation(s)
- Gopal Lamichhane
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (G.L.); (S.-J.L.); (D.-Y.L.)
| | - Jing Liu
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (J.L.); (G.Z.)
| | - Su-Jeong Lee
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (G.L.); (S.-J.L.); (D.-Y.L.)
| | - Da-Yeon Lee
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (G.L.); (S.-J.L.); (D.-Y.L.)
| | - Guolong Zhang
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (J.L.); (G.Z.)
| | - Yoo Kim
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (G.L.); (S.-J.L.); (D.-Y.L.)
| |
Collapse
|
8
|
Moon DO. NADPH Dynamics: Linking Insulin Resistance and β-Cells Ferroptosis in Diabetes Mellitus. Int J Mol Sci 2023; 25:342. [PMID: 38203517 PMCID: PMC10779351 DOI: 10.3390/ijms25010342] [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: 11/15/2023] [Revised: 12/20/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
This review offers an in-depth exploration of Nicotinamide Adenine Dinucleotide Phosphate (NADPH) in metabolic health. It delves into how NADPH affects insulin secretion, influences insulin resistance, and plays a role in ferroptosis. NADPH, a critical cofactor in cellular antioxidant systems and lipid synthesis, plays a central role in maintaining metabolic homeostasis. In adipocytes and skeletal muscle, NADPH influences the pathophysiology of insulin resistance, a hallmark of metabolic disorders such as type 2 diabetes and obesity. The review explores the mechanisms by which NADPH contributes to or mitigates insulin resistance, including its role in lipid and reactive oxygen species (ROS) metabolism. Parallelly, the paper investigates the dual nature of NADPH in the context of pancreatic β-cell health, particularly in its relation to ferroptosis, an iron-dependent form of programmed cell death. While NADPH's antioxidative properties are crucial for preventing oxidative damage in β-cells, its involvement in lipid metabolism can potentiate ferroptotic pathways under certain pathological conditions. This complex relationship underscores the delicate balance of NADPH homeostasis in pancreatic health and diabetes pathogenesis. By integrating findings from recent studies, this review aims to illuminate the nuanced roles of NADPH in different tissues and its potential as a therapeutic target. Understanding these dynamics offers vital insights into the development of more effective strategies for managing insulin resistance and preserving pancreatic β-cell function, thereby advancing the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Dong-Oh Moon
- Department of Biology Education, Daegu University, 201 Daegudae-ro, Gyeongsan-si 38453, Gyeongsangbuk-do, Republic of Korea
| |
Collapse
|
9
|
Park YJ, Kim HY, Shin S, Lee J, Heo I, Cha YY, An HJ. Anti-obesity effect of Lythri herba water extracts in vitro and in vivo. JOURNAL OF ETHNOPHARMACOLOGY 2023; 317:116789. [PMID: 37328083 DOI: 10.1016/j.jep.2023.116789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 06/07/2023] [Accepted: 06/13/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Lythrum salicaria L., also called purple loosestrife, has traditionally been used as a medicinal plant to treat internal dysfunction, such as gastrointestinal disorders or hemorrhages. It contains numerous phytochemical compounds, including orientin, and has been reported to have anti-diarrheal, anti-inflammatory, antioxidant, and antimicrobial properties. AIM OF THE STUDY The effects of Lythrum salicaria L. on obesity have not been explored. Therefore, we investigated the anti-obesity effects of Lythri Herba, the aerial part of this plant, in vitro and in vivo. MATERIALS AND METHODS Using distilled water, Lythri Herba water extracts (LHWE) were prepared by extracting Lythri Herba at 100°Ϲ. The contents of orientin in LHWE were identified using High Performance Liquid Chromatography (HPLC) analysis. To evaluate the anti-obesity effect of LHWE, 3T3-L1 adipocytes and a high-fat diet (HFD)-fed mice were used. Oil-red O staining was performed to examine the anti-adipogenic effects of LHWE in vitro. The histological changes in epididymal white adipose tissue (epiWAT) by LHWE were examined using hematoxylin and eosin staining. Serum leptin levels were measured by enzyme-linked immunosorbent assay. Specific quantification kits measured total cholesterol and triglyceride levels in the serum. The relative fold induction of protein and mRNA was determined using western blot and Quantitative real-time Polymerase Chain Reaction analysis, respectively. RESULTS HPLC analysis demonstrated the presence of orientin in LHWE. LHWE treatment markedly reduced lipid accumulation in differentiated 3T3-L1 adipocytes. LHWE administration also conferred resistance to HFD-induced weight gain in mice and reduced epiWAT mass. Mechanistically, LHWE significantly decreased lipogenesis by downregulating lipoprotein lipase (LPL), glucose-6-phosphate dehydrogenase, ATP-citrate lyase, fatty acid synthase, stearoyl-CoA desaturase 1, sterol regulatory element binding transcription factor 1, and carbohydrate response element binding protein expression and increased the expression of genes involved in fatty acid oxidation (FAO), peroxisome proliferator-activated receptor α and carnitine palmitoyltransferase 1 in 3T3-L1 adipocytes and epiWAT. Furthermore, LHWE significantly up-regulated the phosphorylation of AMP-activated protein kinase in 3T3-L1 adipocytes and epiWAT. CONCLUSION LHWE decreases white adipogenesis in vitro and HFD-induced weight gain in vivo, which is associated with reduced lipogenesis and enhanced FAO.
Collapse
Affiliation(s)
- Yea-Jin Park
- Department of Rehabilitative Medicine of Korean Medicine and Neuropsychiatry, College of Korean Medicine, Sangji University, Wonju, Gangwon-do, 26339, Republic of Korea; Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Hee-Young Kim
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea; Department of Integrated Drug Development and Natural Products, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Su Shin
- Research Institute, BIO PORT KOREA INC., 36, Ballyongsandan 1-ro, Jangan-eup, Gijang-gun, Busan, 46034, Republic of Korea.
| | - JungHyun Lee
- Research Institute, BIO PORT KOREA INC., 36, Ballyongsandan 1-ro, Jangan-eup, Gijang-gun, Busan, 46034, Republic of Korea.
| | - In Heo
- School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, Republic of Korea; Department of Rehabilitation Medicine of Korean Medicine, Pusan National University Korean Medicine Hospital, Yangsan, Gyeongsangnam-do, Republic of Korea.
| | - Yun-Yeop Cha
- Department of Rehabilitative Medicine of Korean Medicine and Neuropsychiatry, College of Korean Medicine, Sangji University, Wonju, Gangwon-do, 26339, Republic of Korea.
| | - Hyo-Jin An
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea; Department of Integrated Drug Development and Natural Products, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
10
|
Davies R, Minhas S, Jayasena CN. The role of seminal reactive oxygen species assessment in the setting of infertility and early pregnancy loss. World J Urol 2023; 41:3257-3265. [PMID: 37452867 PMCID: PMC10632302 DOI: 10.1007/s00345-023-04472-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/31/2023] [Indexed: 07/18/2023] Open
Abstract
The male contribution to a couple suffering with adverse early pregnancy outcomes is being increasingly investigated. Seminal oxidative stress is considered to cause sperm DNA damage, thus affecting the functional capacity of the sperm. Multiple lines of evidence support an association between elevated seminal reactive oxygen species (ROS) and infertility. In the setting of assisted reproduction various factors in the in vitro environment, differing from the in vivo environment, may exacerbate oxidative stress. Furthermore, seminal ROS levels have been found to be higher in the male partners of couple's affected by both spontaneous and recurrent pregnancy loss. There are several methods by which to assess ROS levels however they are costly, inconsistent and their incorporation into clinical practice is unclear. The value of ROS assessment lies in the ability to plan targeted therapies to improve pregnancy and live birth rates. As such, further robust study is required before firm conclusions can be made to inform clinical practice. We aim to review the available evidence regarding the role of seminal ROS in infertility and pregnancy loss.
Collapse
Affiliation(s)
- Rhianna Davies
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Suks Minhas
- Department of Urology, Charing Cross Hospital, Imperial College NHS Trust, London, UK
| | - Channa N Jayasena
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| |
Collapse
|
11
|
Zhang J, Keibler MA, Dong W, Ghelfi J, Cordes T, Kanashova T, Pailot A, Linster CL, Dittmar G, Metallo CM, Lautenschlaeger T, Hiller K, Stephanopoulos G. Stable Isotope-Assisted Untargeted Metabolomics Identifies ALDH1A1-Driven Erythronate Accumulation in Lung Cancer Cells. Biomedicines 2023; 11:2842. [PMID: 37893215 PMCID: PMC10604529 DOI: 10.3390/biomedicines11102842] [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: 08/07/2023] [Revised: 10/08/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Using an untargeted stable isotope-assisted metabolomics approach, we identify erythronate as a metabolite that accumulates in several human cancer cell lines. Erythronate has been reported to be a detoxification product derived from off-target glycolytic metabolism. We use chemical inhibitors and genetic silencing to define the pentose phosphate pathway intermediate erythrose 4-phosphate (E4P) as the starting substrate for erythronate production. However, following enzyme assay-coupled protein fractionation and subsequent proteomics analysis, we identify aldehyde dehydrogenase 1A1 (ALDH1A1) as the predominant contributor to erythrose oxidation to erythronate in cell extracts. Through modulating ALDH1A1 expression in cancer cell lines, we provide additional support. We hence describe a possible alternative route to erythronate production involving the dephosphorylation of E4P to form erythrose, followed by its oxidation by ALDH1A1. Finally, we measure increased erythronate concentrations in tumors relative to adjacent normal tissues from lung cancer patients. These findings suggest the accumulation of erythronate to be an example of metabolic reprogramming in cancer cells, raising the possibility that elevated levels of erythronate may serve as a biomarker of certain types of cancer.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; (J.Z.); (M.A.K.); (W.D.)
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4367 Belvaux, Luxembourg (A.P.)
- Biomia Aps, Kemitorvet 220, 2800 Kongens Lyngby, Denmark
| | - Mark A. Keibler
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; (J.Z.); (M.A.K.); (W.D.)
- Alnylam Pharmaceuticals, Cambridge, MA 02139, USA
| | - Wentao Dong
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; (J.Z.); (M.A.K.); (W.D.)
- Department of Chemical Engineering, Department of Genetics, Institute for Chemistry, Engineering & Medicine for Human Health, Stanford University, Stanford, CA 94305, USA
| | - Jenny Ghelfi
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4367 Belvaux, Luxembourg (A.P.)
| | - Thekla Cordes
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4367 Belvaux, Luxembourg (A.P.)
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | - Tamara Kanashova
- Max-Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Arnaud Pailot
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4367 Belvaux, Luxembourg (A.P.)
| | - Carole L. Linster
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4367 Belvaux, Luxembourg (A.P.)
| | - Gunnar Dittmar
- Max-Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
- Luxembourg Institute of Health, L-1445 Strassen, Luxembourg
| | - Christian M. Metallo
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; (J.Z.); (M.A.K.); (W.D.)
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Tim Lautenschlaeger
- Department of Radiation Oncology, Wexner Medical Center, Ohio State University, Columbus, OH 43221, USA
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Karsten Hiller
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4367 Belvaux, Luxembourg (A.P.)
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | - Gregory Stephanopoulos
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; (J.Z.); (M.A.K.); (W.D.)
| |
Collapse
|
12
|
Duan G, Zheng C, Yu J, Zhang P, Wan M, Zheng J, Duan Y. β-Hydroxy-β-methyl Butyrate Regulates the Lipid Metabolism, Mitochondrial Function, and Fat Browning of Adipocytes. Nutrients 2023; 15:nu15112550. [PMID: 37299513 DOI: 10.3390/nu15112550] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 05/24/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
A growing number of in vivo studies demonstrated that β-hydroxy-β-methyl butyrate (HMB) can serve as a lipid-lowering nutrient. Despite this interesting observation, the use of adipocytes as a model for research is yet to be explored. To ascertain the effects of HMB on the lipid metabolism of adipocytes and elucidate the underlying mechanisms, the 3T3-L1 cell line was employed. Firstly, serial doses of HMB were added to 3T3-L1 preadipocytes to evaluate the effects of HMB on cell proliferation. HMB (50 µM) significantly promoted the proliferation of preadipocytes. Next, we investigated whether HMB could attenuate fat accumulation in adipocytes. The results show that HMB treatment (50 µM) reduced the triglyceride (TG) content. Furthermore, HMB was found to inhibit lipid accumulation by suppressing the expression of lipogenic proteins (C/EBPα and PPARγ) and increasing the expression of lipolysis-related proteins (p-AMPK, p-Sirt1, HSL, and UCP3). We also determined the concentrations of several lipid metabolism-related enzymes and fatty acid composition in adipocytes. The HMB-treated cells showed reduced G6PD, LPL, and ATGL concentrations. Moreover, HMB improved the fatty acid composition in adipocytes, manifested by increases in the contents of n6 and n3 PUFAs. The enhancement of the mitochondrial respiratory function of 3T3-L1 adipocytes was confirmed via Seahorse metabolic assay, which showed that HMB treatment elevated basal mitochondrial respiration, ATP production, H+ leak, maximal respiration, and non-mitochondrial respiration. In addition, HMB enhanced fat browning of adipocytes, and this effect might be associated with the activation of the PRDM16/PGC-1α/UCP1 pathway. Taken together, HMB-induced changes in the lipid metabolism and mitochondrial function may contribute to preventing fat deposition and improving insulin sensitivity.
Collapse
Affiliation(s)
- Geyan Duan
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Changbing Zheng
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Jiayi Yu
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Peiwen Zhang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Mengliao Wan
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Jie Zheng
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yehui Duan
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
13
|
Davies R, Jayasena CN, Rai R, Minhas S. The Role of Seminal Oxidative Stress in Recurrent Pregnancy Loss. Antioxidants (Basel) 2023; 12:antiox12030723. [PMID: 36978971 PMCID: PMC10045000 DOI: 10.3390/antiox12030723] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/01/2023] [Accepted: 03/07/2023] [Indexed: 03/17/2023] Open
Abstract
Recurrent pregnancy loss is a distressing condition affecting 1–2% of couples. Traditionally investigations have focused on the female, however more recently researchers have started to explore the potential contribution of the male partner. Seminal reactive oxygen species have a physiological function in male reproduction but in excess are suspected to generate structural and functional damage to the sperm. Evidence is mounting to support an association between elevated seminal reaction oxygen species and recurrent pregnancy loss. Studies suggest that the rates of sperm DNA damage are higher in the male partners of women affected by recurrent pregnancy loss compared with unaffected men. However, the available pool of data is conflicting, and interpretation is limited by the recent change in nomenclature and the heterogeneity of study methodologies. Furthermore, investigation into the effects of oxidative stress on the epigenome show promise. The value of antioxidant therapy in the management of recurrent pregnancy loss currently remains unclear.
Collapse
Affiliation(s)
- Rhianna Davies
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, UK
| | - Channa N. Jayasena
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, UK
| | - Raj Rai
- Department of Obstetrics and Gynaecology, St Mary’s Hospital, Imperial College NHS Trust, London W2 1NY, UK
| | - Suks Minhas
- Department of Urology, Charing Cross Hospital, Imperial College NHS Trust, London W6 8RF, UK
- Correspondence:
| |
Collapse
|
14
|
Aydemir D, Ulusu NN. The impact of the endocrine-disrupting chemicals on the glucose-6-phosphate dehydrogenase enzyme activity. Front Pharmacol 2023; 14:1133741. [PMID: 36992836 PMCID: PMC10040789 DOI: 10.3389/fphar.2023.1133741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/01/2023] [Indexed: 03/14/2023] Open
Affiliation(s)
- Duygu Aydemir
- Department of Medical Biochemistry, School of Medicine, Koc University, Sariyer, Istanbul, Turkey
- Koç University Research Center for Translational Medicine (KUTTAM), Sariyer, Istanbul, Turkey
| | - Nuriye Nuray Ulusu
- Department of Medical Biochemistry, School of Medicine, Koc University, Sariyer, Istanbul, Turkey
- Koç University Research Center for Translational Medicine (KUTTAM), Sariyer, Istanbul, Turkey
- *Correspondence: Nuriye Nuray Ulusu,
| |
Collapse
|
15
|
More Than an Antioxidant: Role of Dietary Astaxanthin on Lipid and Glucose Metabolism in the Liver of Rainbow Trout ( Oncorhynchus mykiss). Antioxidants (Basel) 2023; 12:antiox12010136. [PMID: 36670998 PMCID: PMC9854815 DOI: 10.3390/antiox12010136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/26/2022] [Accepted: 01/04/2023] [Indexed: 01/09/2023] Open
Abstract
This study investigated the influence of dietary astaxanthin (AX) on glucose and lipid metabolism in rainbow trout liver. Two iso-nitrogenous and iso-lipidic diets were tested for 12 weeks in rainbow trout with an initial mean weight of 309 g. The S-ASTA diet was supplemented with 100 mg of synthetic AX per kg of feed, whereas the control diet (CTRL) had no AX. Fish fed the S-ASTA diet displayed lower neutral and higher polar lipids in the liver, associated with smaller hepatocytes and lower cytoplasm vacuolization. Dietary AX upregulated adipose triglyceride lipase (atgl), hormone-sensitive lipase (hsl2) and 1,2-diacylglycerol choline phosphotransferase (chpt), and downregulated diacylglycerol acyltransferase (dgat2), suggesting the AX's role in triacylglycerol (TAG) turnover and phospholipid (PL) synthesis. Dietary AX may also affect beta-oxidation with the upregulation of carnitine palmitoyltransferase 1 (cpt1α2). Although hepatic cholesterol levels were not affected, dietary AX increased gene expression of sterol regulatory element-binding protein 2 (srebp2). Dietary AX upregulated the expression of 6-phosphogluconate dehydrogenase (6pgdh) and downregulated pyruvate kinase (pkl). Overall, results suggest that dietary AX modulates the oxidative phase of the pentose phosphate pathway and the last step of glycolysis, affecting TAG turnover, β-oxidation, PL and cholesterol synthesis in rainbow trout liver.
Collapse
|
16
|
Acquired Glucose-6-Phosphate Dehydrogenase Deficiency. J Clin Med 2022; 11:jcm11226689. [PMID: 36431166 PMCID: PMC9695330 DOI: 10.3390/jcm11226689] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 10/25/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Glucose-6-phosphate dehydrogenase (G6PD) deficiency is a hereditary condition caused by mutations on chromosome X and is transmitted by a sex-linked inheritance. However, impairment of G6PD activity may result from biochemical mechanisms that are able to inhibit the enzyme in specific clinical conditions in the absence of a structural gene-level defect. In this narrative review, a number of clinical settings associated with an "acquired" G6PD deficiency, phenotypically undistinguishable from the primary deficiency, as well as the mechanisms involved, were examined. Hyperaldosteronism and diabetes are the most common culprits of acquired G6PD deficiency. Additional endocrine and metabolic conditions may cause G6PD deficiency in both hospitalized and outpatients. Contrary to the inherited defect, acquired G6PD deficiency is a condition that is potentially curable by removing the factor responsible for enzyme inhibition. Awareness regarding acquired G6PD deficiency by physicians might result in improved recognition and treatment.
Collapse
|
17
|
Rodríguez-González GL, Vargas-Hernández L, Reyes-Castro LA, Ibáñez CA, Bautista CJ, Lomas-Soria C, Itani N, Estrada-Gutierrez G, Espejel-Nuñez A, Flores-Pliego A, Montoya-Estrada A, Reyes-Muñoz E, Taylor PD, Nathanielsz PW, Zambrano E. Resveratrol Supplementation in Obese Pregnant Rats Improves Maternal Metabolism and Prevents Increased Placental Oxidative Stress. Antioxidants (Basel) 2022; 11:antiox11101871. [PMID: 36290594 PMCID: PMC9598144 DOI: 10.3390/antiox11101871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 11/24/2022] Open
Abstract
Maternal obesity (MO) causes maternal and fetal oxidative stress (OS) and metabolic dysfunction. We investigated whether supplementing obese mothers with resveratrol improves maternal metabolic alterations and reduces OS in the placenta and maternal and fetal liver. From weaning through pregnancy female Wistar rats ate chow (C) or a high-fat diet (MO). One month before mating until 19 days’ gestation (dG), half the rats received 20 mg resveratrol/kg/d orally (Cres and MOres). At 19dG, maternal body weight, retroperitoneal fat adipocyte size, metabolic parameters, and OS biomarkers in the placenta and liver were determined. MO mothers showed higher body weight, triglycerides and leptin serum concentrations, insulin resistance (IR), decreased small and increased large adipocytes, liver fat accumulation, and hepatic upregulation of genes related to IR and inflammatory processes. Placenta, maternal and fetal liver OS biomarkers were augmented in MO. MOres mothers showed more small and fewer large adipocytes, lower triglycerides serum concentrations, IR and liver fat accumulation, downregulation of genes related to IR and inflammatory processes, and lowered OS in mothers, placentas, and female fetal liver. Maternal resveratrol supplementation in obese rats improves maternal metabolism and reduces placental and liver OS of mothers and fetuses in a sex-dependent manner.
Collapse
Affiliation(s)
- Guadalupe L. Rodríguez-González
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - Lilia Vargas-Hernández
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
- Hospital de Ginecología y Obstetricia No. 4 Luis Castelazo Ayala, Mexico City 01090, Mexico
| | - Luis A. Reyes-Castro
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - Carlos A. Ibáñez
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - Claudia J. Bautista
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - Consuelo Lomas-Soria
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
- CONACyT-Cátedras, Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - Nozomi Itani
- Department of Women and Children’s Health, School of Life Course and Population Sciences, King’s College London and King’s Health Partners, London SE1 7EH, UK
| | - Guadalupe Estrada-Gutierrez
- Research Direction, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | - Aurora Espejel-Nuñez
- Department of Immunobiochemistry, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | - Arturo Flores-Pliego
- Department of Immunobiochemistry, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | - Araceli Montoya-Estrada
- Coordination of Gynecological and Perinatal Endocrinology, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | - Enrique Reyes-Muñoz
- Coordination of Gynecological and Perinatal Endocrinology, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | - Paul D. Taylor
- Department of Women and Children’s Health, School of Life Course and Population Sciences, King’s College London and King’s Health Partners, London SE1 7EH, UK
| | - Peter W. Nathanielsz
- Wyoming Center for Pregnancy and Life Course Health Research, Department of Animal Science, University of Wyoming, Laramie, WY 82071, USA
| | - Elena Zambrano
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
- Correspondence: ; Tel.: +52-55-5487-0900 (ext. 2417)
| |
Collapse
|
18
|
Cominetti O, Núñez Galindo A, Corthésy J, Carayol J, Germain N, Galusca B, Estour B, Hager J, Gheldof N, Dayon L. Proteomics reveals unique plasma signatures in constitutional thinness. Proteomics Clin Appl 2022; 16:e2100114. [PMID: 35579096 PMCID: PMC9787820 DOI: 10.1002/prca.202100114] [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: 11/11/2021] [Revised: 04/14/2022] [Accepted: 05/13/2022] [Indexed: 12/30/2022]
Abstract
PURPOSE Studying the plasma proteome of control versus constitutionally thin (CT) individuals, exposed to overfeeding, may give insights into weight-gain management, providing relevant information to the clinical entity of weight-gain resistant CT, and discovering new markers for the condition. EXPERIMENTAL DESIGN Untargeted protein relative quantification of 63 CT and normal-weight individuals was obtained in blood plasma at baseline, during and after an overfeeding challenge using mass spectrometry-based proteomics. RESULTS The plasma proteome of CT subjects presented limited specificity with respect to controls at baseline. Yet, CT showed lower levels of inflammatory C-reactive protein and larger levels of protective insulin-like growth factor-binding protein 2. Differences were more marked during and after overfeeding. CT plasma proteome showed larger magnitude and significance in response, suggesting enhanced "resilience" and more rapid adaptation to changes. Four proteins behaved similarly between CT and controls, while five were regulated in opposite fashion. Ten proteins were differential during overfeeding in CT only (including increased fatty acid-binding protein and glyceraldehyde-3-phosphate dehydrogenase, and decreased apolipoprotein C-II and transferrin receptor protein 1). CONCLUSIONS AND CLINICAL RELEVANCE This first proteomic profiling of a CT cohort reveals different plasma proteomes between CT subjects and controls in a longitudinal clinical trial. Our molecular observations further support that the resistance to weight gain in CT subjects appears predominantly biological. CLINICALTRIALS gov Identifier: NCT02004821.
Collapse
Affiliation(s)
- Ornella Cominetti
- Nestlé Institute of Food Safety & Analytical SciencesNestlé ResearchLausanneSwitzerland
| | - Antonio Núñez Galindo
- Nestlé Institute of Food Safety & Analytical SciencesNestlé ResearchLausanneSwitzerland
| | - John Corthésy
- Nestlé Institute of Food Safety & Analytical SciencesNestlé ResearchLausanneSwitzerland
| | - Jérôme Carayol
- Nestlé Institute of Health SciencesNestlé ResearchLausanneSwitzerland,Present address:
Playtika Switzerland SARue du Port‐Franc 2ALausanne1003Switzerland
| | - Natacha Germain
- Division of EndocrinologyDiabetes, Metabolism and Eating Disorders, CHU St‐EtienneFrance
| | - Bogdan Galusca
- Division of EndocrinologyDiabetes, Metabolism and Eating Disorders, CHU St‐EtienneFrance
| | - Bruno Estour
- Division of EndocrinologyDiabetes, Metabolism and Eating Disorders, CHU St‐EtienneFrance
| | - Jörg Hager
- Nestlé Institute of Health SciencesNestlé ResearchLausanneSwitzerland
| | - Nele Gheldof
- Nestlé Institute of Health SciencesNestlé ResearchLausanneSwitzerland,Present address:
VPA ‐ AVP‐R‐Administration, EPFLBI A2 483, Station 7Lausanne1015Switzerland
| | - Loïc Dayon
- Nestlé Institute of Food Safety & Analytical SciencesNestlé ResearchLausanneSwitzerland,Institut des Sciences et Ingénierie ChimiquesÉcole Polytechnique Fédérale de Lausanne (EPFL)LausanneSwitzerland
| |
Collapse
|
19
|
Insulin sensitivity is associated with the observed variation of de novo lipid synthesis and body composition in finishing pigs. Sci Rep 2022; 12:14586. [PMID: 36028540 PMCID: PMC9418310 DOI: 10.1038/s41598-022-18799-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 08/19/2022] [Indexed: 11/08/2022] Open
Abstract
Variations in body composition among pigs can be associated with insulin sensitivity given the insulin anabolic effect. The study objectives were to characterize this association and to compare de novo lipogenesis and the gene expression in the adipose tissue of pigs of the same genetic background. Thirty 30-95 kg of body weight (BW) pigs, catheterized in the jugular vein participated into an oral glucose tolerance test (OGTT; 1.75 g glucose/kg of BW) to calculate insulin-related indexes. The 8 fattest and the 8 leanest pigs were used to determine the relative mRNA abundance of studied genes. The rate of lipogenesis was assessed by incorporation of [U-13C]glucose into lipids. The QUICKI and Matsuda indexes negatively correlated with total body lipids (r = - 0.67 and r = - 0.59; P < 0.01) and de novo lipogenesis (r = - 0.58; P < 0.01). Fat pigs had a higher expression level of lipogenic enzymes (ACACA, ACLY; P < 0.05) than lean pigs. The reduced insulin sensitivity in fat pigs was associated with a higher expression level of glucose-6-phosphate dehydrogenase (G6PD) and a lower expression of peroxisome proliferator-activated receptor-gamma (PPAR-γ). In conclusion, pigs with increased body lipids have lower insulin sensitivity which is associated with increased de novo lipogenesis.
Collapse
|
20
|
Zhang S, Kong L, Jia Z, Shao S, Pan L, Wang W, Sun Y. Anti-obesity effects of corn peptide on 3T3-L1 preadipocytes and C57BL/6J obese mice. Arch Anim Nutr 2022; 76:205-220. [PMID: 36645054 DOI: 10.1080/1745039x.2022.2162800] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 12/15/2022] [Indexed: 01/17/2023]
Abstract
Corn peptide (CP) is a small, natural, biologically active peptide obtained by protease-catalysed hydrolysis of corn. CP exerts antihypertensive, hypoglycaemic, antihyperlipidemic, antioxidant, and antitumor effects, as well as prevents cardiovascular and cerebrovascular diseases. Although CP plays a role in preventing obesity-related diseases, its role in reducing obesity has not yet been determined. In this study, we analysed the inhibitory effects of CP on lipid droplet accumulation in 3T3-L1 preadipocytes and high-fat diet (HFD)-induced C57BL/6J Obese Mice. The results show that CP could inhibit preadipocyte differentiation and oil accumulation in 3T3-L1 preadipocytes. Oral CP administration reduced serum triglyceride (TG) content, epididymal fat weight, abnormal liver fat droplet accumulation, and C/EBPα expression. Furthermore, combination of CP administration and exercise reduced body, liver, and adipose tissue weights; decreased serum total cholesterol (TC), triglyceride and low-density lipoprotein (LDL) levels; and inhibited hepatic lipid droplet accumulations and epididymal fat cell hypertrophy. Additionally, this combination inhibited the expression of transcription factors, C/EBPα, C/EBPβ, and PPARγ, and adipogenic factors, FABP4 in mice. In conclusion, oral administration of CP inhibited lipid droplet accumulation and counteracted HFD-induced obesity in mice.
Collapse
Affiliation(s)
- Shanshan Zhang
- College of Life Science and Agriculture Forestry, Qiqihar University, Qiqihar, China
| | - Lingzhe Kong
- College of Life Science and Agriculture Forestry, Qiqihar University, Qiqihar, China
| | | | - Shuli Shao
- College of Life Science and Agriculture Forestry, Qiqihar University, Qiqihar, China
| | - Lin Pan
- College of Life Science and Agriculture Forestry, Qiqihar University, Qiqihar, China
| | - Weiyu Wang
- College of Life Science and Agriculture Forestry, Qiqihar University, Qiqihar, China
| | - Yingning Sun
- College of Life Science and Agriculture Forestry, Qiqihar University, Qiqihar, China
| |
Collapse
|
21
|
Anti-Obesity Effects of a Mixture of Atractylodes macrocephala and Amomum villosum Extracts on 3T3-L1 Adipocytes and High-Fat Diet-Induced Obesity in Mice. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27030906. [PMID: 35164174 PMCID: PMC8839705 DOI: 10.3390/molecules27030906] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/26/2022] [Accepted: 01/26/2022] [Indexed: 01/13/2023]
Abstract
Since the potential of (3:1) mixtures of Atractylodes macrocephala and Amomum villosum extracts has been proposed in the management of obesity, the purpose of present study was to investigate the effects of AME:AVE (3:1) mixture on weight loss, obesity-related biochemical parameters, adipogenesis and lipogenesis related proteins in 3T3-L1 cells and HFD-induced obesity in a mouse model. Treatment with AME:AVE (3:1) mixture inhibited lipid accumulation. Furthermore, the treatment with 75 and 150 mg/kg of AME:AVE (3:1) significantly decreased the body weight gain, white adipose tissue (WAT) weight, and plasma glucose level in HFD-induced obese mice. Moreover, treatment with 75 and 150 mg/kg AME:AVE (3:1) also significantly lowered the size of adipocytes in adipose tissue and reduced the lipid accumulation in liver. AME:AVE (3:1) treatment significantly decreased the expression of proteins related to adipogenesis and lipogenesis in 3T3-L1 adipocytes and WAT of HFD-induced obese mice. These results suggest that the AME:AVE herbal mixture (3:1) has anti-obesity effects, which may be elicited by regulating the expression of adipogenesis and lipogenesis-related proteins in adipocytes and WAT in HFD-induced obesity in mice.
Collapse
|
22
|
Hu Y, Cai M, Zhong H, Chu W, Hu Y. A Study on How Methionine Restriction Decreases the Body's Hepatic and Lipid Deposition in Rice Field Eel ( Monopterus albus). Int J Mol Sci 2021; 22:ijms222413379. [PMID: 34948174 PMCID: PMC8705440 DOI: 10.3390/ijms222413379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 11/16/2022] Open
Abstract
Methionine restriction reduces animal lipid deposition. However, the molecular mechanism underlying how the body reacts to the condition and regulates lipid metabolism remains unknown. In this study, a feeding trial was performed on rice field eel Monopterus albus with six isonitrogenous and isoenergetic feeds that included different levels of methionine (0, 2, 4, 6, 8, and 10 g/kg). Compared with M0 (0 g/kg), the crude lipid and crude protein of M. albus increased markedly in M8 (8 g/kg) (p < 0.05), serum (total cholesterol, triglyceride, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, and non-esterified free fatty acids), and hepatic contents (hepatic lipase, apolipoprotein-A, fatty acid synthetase, total cholesterol, triglyceride, and lipoprteinlipase). However, in the serum, very-low-density lipoprotein and hepatic contents (hormone-sensitive triglyceride lipase, Acetyl CoA carboxylase, carnitine palmitoyltransterase, and mirosomal triglygeride transfer protein) decreased markedly in M8 (p < 0.05). The contents of hepatic C18:2n-6, C22:6n-3, and n-3PUFA in the M8 group were significantly higher than those in M0 (p < 0.05), and the contents of lipid droplets in M8 were higher than those in M0. Compared with M0, the hepatic gcn2, eif2α, hsl, mttp, ldlrap, pparα, cpt1, and cpt2 were remarkably downregulated in M8, while srebf2, lpl, moat2, dgat2, hdlbp, srebf1, fas, fads2, me1, pfae, and icdh were markedly upregulated in M8. Moreover, hepatic SREBP1 and FAS protein expression were upregulated significantly in M8 (p < 0.01). In short, methionine restriction decreased the lipid deposition of M. albus, especially for hepatic lipid deposition, and mainly downregulated hepatic fatty acid metabolism. Besides, gcn2 could be activated under methionine restriction.
Collapse
Affiliation(s)
- Yajun Hu
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha 410128, China; (Y.H.); (M.C.); (H.Z.)
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Minglang Cai
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha 410128, China; (Y.H.); (M.C.); (H.Z.)
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Huan Zhong
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha 410128, China; (Y.H.); (M.C.); (H.Z.)
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Wuying Chu
- Department of Bioengineering and Environmental Science, Changsha University, Changsha 410000, China;
| | - Yi Hu
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha 410128, China; (Y.H.); (M.C.); (H.Z.)
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Correspondence:
| |
Collapse
|
23
|
Park S, Lee JJ, Shin HW, Jung S, Ha JH. Effect of Soybean and Soybean Koji on Obesity and Dyslipidemia in Rats Fed a High-Fat Diet: A Comparative Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:6032. [PMID: 34199728 PMCID: PMC8199977 DOI: 10.3390/ijerph18116032] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 12/19/2022]
Abstract
Soybean koji refers to steamed soybeans inoculated with microbial species. Soybean fermentation improves the health benefits of soybeans. Obesity is a serious health concern owing to its increasing incidence rate and high association with other metabolic diseases. Therefore, we investigated the effects of soybean and soybean koji on high-fat diet-induced obesity in rats. Five-week-old male Sprague-Dawley rats were randomly divided into four groups (n = 8/group) as follows: (1) regular diet (RD), (2) high-fat diet (HFD), (3) HFD + steamed soybean (HFD+SS), and (4) HFD + soybean koji (HFD+SK). SK contained more free amino acids and unsaturated fatty acids than SS. In a rat model of obesity, SK consumption significantly alleviated the increase in weight of white adipose tissue and mRNA expression of lipogenic genes, whereas SS consumption did not. Both SS and SK reduced serum triglyceride, total cholesterol, and low-density lipoprotein cholesterol levels, and increased high-density lipoprotein cholesterol levels. SS and SK also inhibited lipid accumulation in the liver and white adipose tissue and reduced adipocyte size. Although both SS and SK could alleviate HFD-induced dyslipidemia, SK has better anti-obesity effects than SS by regulating lipogenesis. Overall, SK is an excellent functional food that may prevent obesity.
Collapse
Affiliation(s)
- Sihoon Park
- Department of Food and Nutrition, Chosun University, Gwangju 61452, Korea; (S.P.); (J.-J.L.)
| | - Jae-Joon Lee
- Department of Food and Nutrition, Chosun University, Gwangju 61452, Korea; (S.P.); (J.-J.L.)
| | - Hye-Won Shin
- Food R&D Institute, CJ CheilJedang Corp., Suwon 16495, Korea;
| | - Sunyoon Jung
- Research Center for Industrialization of Natural Neutralization, Dankook University, Cheonan 31116, Korea
- Department of Food Science and Nutrition, Dankook University, Cheonan 31116, Korea
| | - Jung-Heun Ha
- Research Center for Industrialization of Natural Neutralization, Dankook University, Cheonan 31116, Korea
- Department of Food Science and Nutrition, Dankook University, Cheonan 31116, Korea
| |
Collapse
|
24
|
Sigmarsdottir TB, McGarrity S, Yurkovich JT, Rolfsson Ó, Sigurjónsson ÓE. Analyzing Metabolic States of Adipogenic and Osteogenic Differentiation in Human Mesenchymal Stem Cells via Genome Scale Metabolic Model Reconstruction. Front Cell Dev Biol 2021; 9:642681. [PMID: 34150750 PMCID: PMC8212021 DOI: 10.3389/fcell.2021.642681] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/29/2021] [Indexed: 01/14/2023] Open
Abstract
Since their initial discovery in 1976, mesenchymal stem cells (MSCs) have been gathering interest as a possible tool to further the development and enhancement of various therapeutics within regenerative medicine. However, our current understanding of both metabolic function and existing differences within the varying cell lineages (e.g., cells in either osteogenesis or adipogenesis) is severely lacking making it more difficult to fully realize the therapeutic potential of MSCs. Here, we reconstruct the MSC metabolic network to understand the activity of various metabolic pathways and compare their usage under different conditions and use these models to perform experimental design. We present three new genome-scale metabolic models (GEMs) each representing a different MSC lineage (proliferation, osteogenesis, and adipogenesis) that are biologically feasible and have distinctive cell lineage characteristics that can be used to explore metabolic function and increase our understanding of these phenotypes. We present the most distinctive differences between these lineages when it comes to enriched metabolic subsystems and propose a possible osteogenic enhancer. Taken together, we hope these mechanistic models will aid in the understanding and therapeutic potential of MSCs.
Collapse
Affiliation(s)
| | - Sarah McGarrity
- School of Science and Engineering, Reykjavík University, Reykjavík, Iceland.,Center for Systems Biology, University of Iceland, Reykjavík, Iceland
| | - James T Yurkovich
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
| | - Óttar Rolfsson
- Center for Systems Biology, University of Iceland, Reykjavík, Iceland
| | - Ólafur Eysteinn Sigurjónsson
- School of Science and Engineering, Reykjavík University, Reykjavík, Iceland.,The Blood Bank, Landspitali - The National University Hospital of Iceland, Reykjavík, Iceland
| |
Collapse
|
25
|
Jin SC, Kim MH, Choi LY, Nam YK, Yang WM. Fat regulatory mechanisms of pine nut oil based on protein interaction network analysis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 86:153557. [PMID: 33852976 DOI: 10.1016/j.phymed.2021.153557] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/12/2021] [Accepted: 03/23/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Pine nut oil (PNO), a standardized and well-defined extract of Pinus koraiensis (Korean pine), has beneficial effects on wound healing, inflammatory diseases, and cancer. However, the explanation for the mechanism by which PNO reduces body fat remains uncertain. We performed a protein-protein interaction network (PPIN) analysis to explore the genes associated with pinolenic acid using the MEDILINE database from PubChem and PubMed. It was concluded through the PPIN analysis that PNO was involved in a neutral lipid biosynthetic process. PURPOSE This study evaluated the effects of PNO predicted by the network analysis of fat accumulation in chronic obesity mouse models established by feeding a high fat diet (HFD) to C57BL/6J mice and explored potential mechanisms. METHODS HFD mice were fed only HFD or HFD with PNO at 822 and 1644 mg/kg. After an oral administration of 7 weeks, several body weight and body fat-related parameters were examined, including the following: adipose weight, adipocyte size, serum lipid profiles, adipocyte expression of PPAR-γ, sterol regulatory element binding protein (SREBP)-1c, lipoprotein lipase (LPL) and leptin. RESULTS We showed that oral administration of PNO to HFD mice reduces body fat weight, fat in tissue, white adipose tissue weight, and adipocyte size. The serum cholesterol was improved in the HFD mice treated with PNO. Additionally, PNO has significantly attenuated the HFD-induced changes in the adipose tissue expression of PPAR-γ, SREBP-1c, LPL, and leptin. CONCLUSIONS The findings from this study based on the PPIN analysis suggest that PNO has potential as drug to reduce body fat through fat regulatory mechanisms by PPAR-γ and SREBP-1c.
Collapse
Affiliation(s)
- Seong Chul Jin
- Department of Convergence Korean Medical Science, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Mi Hye Kim
- Department of Convergence Korean Medical Science, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - La Yoon Choi
- Department of Convergence Korean Medical Science, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yeon Kyung Nam
- Department of Convergence Korean Medical Science, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Woong Mo Yang
- Department of Convergence Korean Medical Science, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
26
|
The Controversial Role of Glucose-6-Phosphate Dehydrogenase Deficiency on Cardiovascular Disease: A Narrative Review. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5529256. [PMID: 34007401 PMCID: PMC8110402 DOI: 10.1155/2021/5529256] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/27/2021] [Accepted: 04/21/2021] [Indexed: 12/12/2022]
Abstract
Cardiovascular disorders (CVD) are highly prevalent and the leading cause of death worldwide. Atherosclerosis is responsible for most cases of CVD. The plaque formation and subsequent thrombosis in atherosclerosis constitute an ongoing process that is influenced by numerous risk factors such as hypertension, diabetes, dyslipidemia, obesity, smoking, inflammation, and sedentary lifestyle. Among the various risk and protective factors, the role of glucose-6-phosphate dehydrogenase (G6PD) deficiency, the most common inborn enzyme disorder across populations, is still debated. For decades, it has been considered a protective factor against the development of CVD. However, in the recent years, growing scientific evidence has suggested that this inherited condition may act as a CVD risk factor. The role of G6PD deficiency in the atherogenic process has been investigated using in vitro or ex vivo cellular models, animal models, and epidemiological studies in human cohorts of variable size and across different ethnic groups, with conflicting results. In this review, the impact of G6PD deficiency on CVD was critically reconsidered, taking into account the most recent acquisitions on molecular and biochemical mechanisms, namely, antioxidative mechanisms, glutathione recycling, and nitric oxide production, as well as their mutual interactions, which may be impaired by the enzyme defect in the context of the pentose phosphate pathway. Overall, current evidence supports the notion that G6PD downregulation may favor the onset and evolution of atheroma in subjects at risk of CVD. Given the relatively high frequency of this enzyme deficiency in several regions of the world, this finding might be of practical importance to tailor surveillance guidelines and facilitate risk stratification.
Collapse
|
27
|
Tracking biochemical changes induced by iron loading in AML12 cells with synchrotron live cell, time-lapse infrared microscopy. Biochem J 2021; 478:1227-1239. [PMID: 33616158 DOI: 10.1042/bcj20200653] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 02/01/2021] [Accepted: 02/22/2021] [Indexed: 12/12/2022]
Abstract
Hepatocytes are essential for maintaining the homeostasis of iron and lipid metabolism in mammals. Dysregulation of either iron or lipids has been linked with serious health consequences, including non-alcoholic fatty liver disease (NAFLD). Considered the hepatic manifestation of metabolic syndrome, NAFLD is characterised by dysregulated lipid metabolism leading to a lipid storage phenotype. Mild to moderate increases in hepatic iron have been observed in ∼30% of individuals with NAFLD; however, direct observation of the mechanism behind this increase has remained elusive. To address this issue, we sought to determine the metabolic consequences of iron loading on cellular metabolism using live cell, time-lapse Fourier transform infrared (FTIR) microscopy utilising a synchrotron radiation source to track biochemical changes. The use of synchrotron FTIR is non-destructive and label-free, and allowed observation of spatially resolved, sub-cellular biochemical changes over a period of 8 h. Using this approach, we have demonstrated that iron loading in AML12 cells induced perturbation of lipid metabolism congruent with steatosis development. Iron-loaded cells had approximately three times higher relative ester carbonyl concentration compared with controls, indicating an accumulation of triglycerides. The methylene/methyl ratio qualitatively suggests the acyl chain length of fatty acids in iron-loaded cells increased over the 8 h period of monitoring compared with a reduction observed in the control cells. Our findings provide direct evidence that mild to moderate iron loading in hepatocytes drives de novo lipid synthesis, consistent with a role for iron in the initial hepatic lipid accumulation that leads to the development of hepatic steatosis.
Collapse
|
28
|
Chung SI, Kang MY. Oral Administration of Germinated, Pigmented, Giant Embryo Rice ( Oryza sativa L. cv. Keunnunjami) Extract Improves the Lipid and Glucose Metabolisms in High-Fat Diet-Fed Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8829778. [PMID: 33552386 PMCID: PMC7846407 DOI: 10.1155/2021/8829778] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 12/24/2020] [Accepted: 12/30/2020] [Indexed: 11/18/2022]
Abstract
Obesity is a significant risk factor for chronic diseases. The effect of ethanol extract from germinated Keunnunjami, blackish-purple rice with a giant embryo, compare to ordinary brown rice, on the body weight and lipid and glucose metabolism in high-fat diet-fed mice was analyzed. Mice were fed with a high-fat diet-fed for 3 weeks and then orally administered with either distilled water (HF) or extract (0.25%, w/w) from brown, germinated brown, Keunnunjami, and germinated Keunnunjami rice for 4 weeks. Control mice were fed with a normal diet and orally administered with distilled water. The HF group showed markedly higher body weight and triglyceride, cholesterol, fatty acid, glucose, and insulin levels than the control group. However, the oral administration of rice extracts ameliorated this high-fat diet-induced obesity, hyperlipidemia, and hypoglycemia through the modulation of adipokine production, lipogenic and glucose-regulating enzyme activities, and mRNA expression of genes associated with lipid and glucose metabolism. The germinated Keunnunjami extract exhibited greater hypolipidemic, hypoglycemic, and body weight-lowering effects than the other rice extracts. The results demonstrated that germination could further enhance the physiological properties of rice and that germinated Keunnunjami extract has a strong therapeutic potential against high-fat diet-induced obesity, hyperlipidemia, and hyperglycemia.
Collapse
Affiliation(s)
- Soo Im Chung
- International Agricultural Training Center, Kyungpook National University, Daegu 41566, Republic of Korea
- Department of Food Science and Nutrition, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Mi Young Kang
- Department of Food Science and Nutrition, Kyungpook National University, Daegu 41566, Republic of Korea
| |
Collapse
|
29
|
Choi YE, Choi SI, Han X, Men X, Jang GW, Kwon HY, Kang SR, Han JS, Lee OKH. Radical Scavenging-Linked Anti-Adipogenic Activity of Aster scaber Ethanolic Extract and Its Bioactive Compound. Antioxidants (Basel) 2020; 9:antiox9121290. [PMID: 33339396 PMCID: PMC7766398 DOI: 10.3390/antiox9121290] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 12/13/2022] Open
Abstract
Aster scaber is a wild vegetable cultivated in Korea and is known to contain phytochemicals with various biological activities. The potential antioxidant and anti-obesity effects of A. scaber and their mechanism are yet to be reported. We evaluated the total phenolic, flavonoid, and proanthocyanidin contents and oxygen radical absorbance capacity of A. scaber ethanolic extract (ASE), and analyzed the major phenolic compounds of ASE. Antioxidant activity was measured at the chemical level through 2,2-diphenyl-1-picrylhydrazyl (DPPH), reducing power assay, 2,2′-azino-bis(3-ethylbenzthiazoline-6-sulfonic acid) (ABTS), and fluorescence recovery after photobleaching (FRAP) assay. In addition, it was measured in vitro through inhibition of Reactive oxygen species (ROS) production in 3T3-L1 adipocyte, and inhibition of lipid accumulation was also evaluated. ASE reduced the expression of enzymes involved in the production of ROS and increased the expression of antioxidant enzymes that reduce increased ROS levels. They also reduced the expression of adipogenesis transcription factors that regulate adipocyte differentiation in relation to ROS production, inhibited the expression of lipogenesis-related genes related to fat accumulation through AMP-activated protein kinase (AMPK) activity, and increased expression of lipolysis-related genes. Thus, ASE containing CGA (chlorogenic acid) inhibits ROS production in 3T3-L1 adipocytes, owing to its strong antioxidant activity, and inhibits lipid accumulation caused by oxidative stress. The extract can be used as a potential functional food material for reducing oxidative stress and obesity.
Collapse
Affiliation(s)
- Ye-Eun Choi
- Department of Food Science and Biotechnology, Kangwon National University, Chuncheon 24341, Korea; (Y.-E.C.); (S.-I.C.); (X.H.); (X.M.); (G.-W.J.); (H.-Y.K.)
| | - Sun-Il Choi
- Department of Food Science and Biotechnology, Kangwon National University, Chuncheon 24341, Korea; (Y.-E.C.); (S.-I.C.); (X.H.); (X.M.); (G.-W.J.); (H.-Y.K.)
| | - Xionggao Han
- Department of Food Science and Biotechnology, Kangwon National University, Chuncheon 24341, Korea; (Y.-E.C.); (S.-I.C.); (X.H.); (X.M.); (G.-W.J.); (H.-Y.K.)
| | - Xiao Men
- Department of Food Science and Biotechnology, Kangwon National University, Chuncheon 24341, Korea; (Y.-E.C.); (S.-I.C.); (X.H.); (X.M.); (G.-W.J.); (H.-Y.K.)
| | - Gill-Woong Jang
- Department of Food Science and Biotechnology, Kangwon National University, Chuncheon 24341, Korea; (Y.-E.C.); (S.-I.C.); (X.H.); (X.M.); (G.-W.J.); (H.-Y.K.)
| | - Hee-Yeon Kwon
- Department of Food Science and Biotechnology, Kangwon National University, Chuncheon 24341, Korea; (Y.-E.C.); (S.-I.C.); (X.H.); (X.M.); (G.-W.J.); (H.-Y.K.)
| | - Seong-Ran Kang
- The Food Industry Promotional Agency of Korea, Iksan 54576, Korea; (S.-R.K.); (J.-S.H.)
| | - Jin-Soo Han
- The Food Industry Promotional Agency of Korea, Iksan 54576, Korea; (S.-R.K.); (J.-S.H.)
| | - OK-Hwan Lee
- Department of Food Science and Biotechnology, Kangwon National University, Chuncheon 24341, Korea; (Y.-E.C.); (S.-I.C.); (X.H.); (X.M.); (G.-W.J.); (H.-Y.K.)
- Correspondence: ; Tel.: +82-33-250-6454; Fax: +82-33-259-5565
| |
Collapse
|
30
|
Male Factors: the Role of Sperm in Preimplantation Embryo Quality. Reprod Sci 2020; 28:1788-1811. [DOI: 10.1007/s43032-020-00334-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 09/25/2020] [Indexed: 12/19/2022]
|
31
|
Ortiz SR, Field MS. Mammalian metabolism of erythritol: a predictive biomarker of metabolic dysfunction. Curr Opin Clin Nutr Metab Care 2020; 23:296-301. [PMID: 32412980 DOI: 10.1097/mco.0000000000000665] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW To summarize recent advances in our understanding of mammalian erythritol metabolism and its use as a predictive biomarker of cardiometabolic disease risk. RECENT FINDINGS Elevated serum erythritol predicts future central adiposity gain and type 2 diabetes mellitus in healthy adults. Erythritol is a newly recognized human metabolic product of glucose, synthesized through the pentose phosphate pathway. The final conversion of this metabolic pathway is catalyzed by the enzymes sorbitol dehydrogenase and alcohol dehydrogenase 1. Erythritol is also a well characterized nonnutritive sweetener. Recent studies show that dietary erythritol can be metabolized to erythrose or erythronate in humans before excretion. SUMMARY Elevated serum erythritol predicts risk for cardiometabolic disease, but more research is required to maximize its utility as a biomarker, including characterizing the determinants of endogenous erythritol synthesis from glucose. New insights into dietary erythritol metabolism also highlight the need to evaluate the effects of long-term erythritol consumption.
Collapse
Affiliation(s)
- Semira R Ortiz
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA
| | | |
Collapse
|
32
|
Tian N, Liu Q, Li Y, Tong L, Lu Y, Zhu Y, Zhang P, Chen H, Hu L, Meng J, Feng M, Li M, Zheng L, Li B, Xu T, Wu L, Tong X. Transketolase Deficiency in Adipose Tissues Protects Mice From Diet-Induced Obesity by Promoting Lipolysis. Diabetes 2020; 69:1355-1367. [PMID: 32295803 DOI: 10.2337/db19-1087] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 04/09/2020] [Indexed: 01/06/2023]
Abstract
Obesity has recently become a prevalent health threat worldwide. Although emerging evidence has suggested a strong link between the pentose phosphate pathway (PPP) and obesity, the role of transketolase (TKT), an enzyme in the nonoxidative branch of the PPP that connects PPP and glycolysis, remains obscure in adipose tissues. In this study, we specifically deleted TKT in mouse adipocytes and found no obvious phenotype upon normal diet feeding. However, adipocyte TKT abrogation attenuated high-fat diet-induced obesity, reduced hepatic steatosis, improved glucose tolerance, alleviated insulin resistance, and increased energy expenditure. Mechanistically, TKT deficiency accumulated nonoxidative PPP metabolites and decreased glycolysis and pyruvate input into the mitochondria, leading to increased lipolytic enzyme gene expression and enhanced lipolysis, fatty acid oxidation, and mitochondrial respiration. Therefore, our data not only identify a novel role of TKT in regulating lipolysis and obesity but also suggest that limiting glucose-derived carbon into the mitochondria induces lipid catabolism and energy expenditure.
Collapse
Affiliation(s)
- Na Tian
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yakui Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lingfeng Tong
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Lu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yemin Zhu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping Zhang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hanbei Chen
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Hu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Meng
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Ming Feng
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Minle Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Liang Zheng
- Pediatric Translational Medicine Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Li
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianle Xu
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lifang Wu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuemei Tong
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
33
|
Concepcion J, Chen K, Saito R, Gangoiti J, Mendez E, Nikita ME, Barshop BA, Natarajan L, Sharma K, Kim JJ. Identification of pathognomonic purine synthesis biomarkers by metabolomic profiling of adolescents with obesity and type 2 diabetes. PLoS One 2020; 15:e0234970. [PMID: 32589682 PMCID: PMC7319336 DOI: 10.1371/journal.pone.0234970] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 06/05/2020] [Indexed: 12/18/2022] Open
Abstract
The incidence of type 2 diabetes is increasing more rapidly in adolescents than in any other age group. We identified and compared metabolite signatures in obese children with type 2 diabetes (T2D), obese children without diabetes (OB), and healthy, age- and gender-matched normal weight controls (NW) by measuring 273 analytes in fasting plasma and 24-hour urine samples from 90 subjects by targeted LC-MS/MS. Diabetic subjects were within 2 years of diagnosis in an attempt to capture early-stage disease prior to declining renal function. We found 22 urine metabolites that were uniquely associated with T2D when compared to OB and NW groups. The metabolites most significantly elevated in T2D youth included members of the betaine pathway, nucleic acid metabolism, and branched-chain amino acids (BCAAs) and their catabolites. Notably, the metabolite pattern in OB and T2D groups differed between urine and plasma, suggesting that urinary BCAAs and their intermediates behaved as a more specific biomarker for T2D, while plasma BCAAs associated with the obese, insulin resistant state independent of diabetes status. Correlative analysis of metabolites in the T2D signature indicated that betaine metabolites, BCAAs, and aromatic amino acids were associated with hyperglycemia, but BCAA acylglycine derivatives and nucleic acid metabolites were linked to insulin resistance. Of major interest, we found that urine levels of succinylaminoimidazole carboxamide riboside (SAICA-riboside) were increased in diabetic youth, identifying urine SAICA-riboside as a potential biomarker for T2D.
Collapse
Affiliation(s)
- Jennifer Concepcion
- Department of Pediatrics, University of California San Diego, La Jolla, CA, United States of America
- Rady Children’s Hospital, San Diego, CA, United States of America
| | - Katherine Chen
- Department of Pediatrics, University of California San Diego, La Jolla, CA, United States of America
| | | | - Jon Gangoiti
- Department of Pediatrics, University of California San Diego, La Jolla, CA, United States of America
| | - Eric Mendez
- Department of Pediatrics, University of California San Diego, La Jolla, CA, United States of America
| | - Maria Eleni Nikita
- Department of Pediatrics, University of California San Diego, La Jolla, CA, United States of America
| | - Bruce A. Barshop
- Department of Pediatrics, University of California San Diego, La Jolla, CA, United States of America
- Rady Children’s Hospital, San Diego, CA, United States of America
| | - Loki Natarajan
- Department of Family Medicine and Public Health, University of California San Diego, La Jolla, CA, United States of America
| | - Kumar Sharma
- Center for Renal Precision Medicine, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, Unied States of America
| | - Jane J. Kim
- Department of Pediatrics, University of California San Diego, La Jolla, CA, United States of America
- Rady Children’s Hospital, San Diego, CA, United States of America
- * E-mail:
| |
Collapse
|
34
|
Activity-expression profiling of glucose-6-phosphate dehydrogenase in tissues of normal and diabetic mice. Biochem Biophys Res Commun 2020; 524:750-755. [PMID: 32035617 DOI: 10.1016/j.bbrc.2020.01.165] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 01/30/2020] [Indexed: 10/25/2022]
Abstract
Glucose-6-phosphate dehydrogenase (G6PD) plays a principal role in the regulation of oxidative stress by modulating the nicotinamide adenine dinucleotide phosphate pool and is expected to be associated with metabolic diseases such as diabetes mellitus (DM). However, it is unclear whether hyperglycemia increases G6PD activity levels in DM because suitable assays for quantifying the activity in a high-throughput manner are lacking. Using liquid droplet arrays tailored to analyze tissue lysates, we performed G6PD activity profiling in eight tissues of normal and diabetic mice: brain, heart, kidney, liver, lung, muscle, spleen, and thyroid. Diabetic mice exhibited significantly higher G6PD activities in the kidney, liver, spleen, and thyroid than normal mice; no significant difference was found in the brain, heart, lung, or muscle. We also performed G6PD expression profiling in the eight tissues using Western blot analysis. Diabetic mice showed significantly elevated G6PD expression levels in the kidney, lung, spleen, and thyroid compared with normal mice; no significant difference was found in the brain, heart, liver, or muscle. An analysis of G6PD activity-expression profiles demonstrated tissue-specific changes in response to hyperglycemia. Thus, our approach would be helpful for understanding the role of G6PD in tissue-based pathogenesis of diabetic complications.
Collapse
|
35
|
Palafox MA, Chalanchi SM, Isasi J, Premkumar R, Franklin Benial AM, Rastogi VK. Effect of bromine atom on the different tautomeric forms of microhydrated 5-bromouracil, in the DNA:RNA microhelix and in the interaction with human proteins. J Biomol Struct Dyn 2020; 38:5443-5463. [DOI: 10.1080/07391102.2019.1704878] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- M. Alcolea Palafox
- Facultad de Ciencias Químicas, Departamento de Química-Fisica, Universidad Complutense de Madrid, Madrid, Spain
| | - S. M. Chalanchi
- Department of Chemistry, University of Sistan and Baluchestan, Zahedan, Iran
| | - J. Isasi
- Facultad de Ciencias Químicas, Departamento de Química Inorgánica, Universidad Complutense de Madrid, Madrid, Spain
| | - R. Premkumar
- PG and Research Department of Physics, N.M.S.S.V.N. College, Madurai, Tamil Nadu, India
| | | | | |
Collapse
|
36
|
Sayed S, Ahmed M, El-Shehawi A, Alkafafy M, Al-Otaibi S, El-Sawy H, Farouk S, El-Shazly S. Ginger Water Reduces Body Weight Gain and Improves Energy Expenditure in Rats. Foods 2020; 9:E38. [PMID: 31906567 PMCID: PMC7023345 DOI: 10.3390/foods9010038] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/26/2019] [Accepted: 12/30/2019] [Indexed: 12/22/2022] Open
Abstract
Obesity is a serious global problem that causes predisposition to numerous serious diseases. The current study aims to investigate the effect of ginger water on body weight and energy expenditure through modulation of mRNA expression of carbohydrate and lipid metabolism. A white colored liquid obtained during freeze-drying of fresh rhizomes of Zingiber officinal was collected and named ginger water. It was used to treat rats, then blood and tissue samples were collected from the liver and white adipose at the end of the experiment. The serum was prepared and used for biochemical assays, while tissue samples were used for RNA isolation and gene expression analysis via Reverse transcription polymerase chain reaction (RT-PCR). Results of High Performance Liquid Chromatography (HPLC) analysis of ginger water revealed the presence of chrysin and galangin at concentrations of 0.24 µg/mL and 0.53 µg/mL, respectively. Average body weight gain decreased significantly in groups that received ginger water. In addition, both total cholesterol and serum triacylglycerol were reduced in the groups that received ginger water. Furthermore, mRNA expression of Sterol regulatory element-binding protein 1 (SREBP-1c) in the liver and leptin in adipose tissues were downregulated, while those of adiponectin, hepatic carnitine palmitoyltransferase1 (CPT-1), acyl-coA oxidase (ACO), Glucose transporter 2 (GLUT-2), and pyruvate kinase (PK) were upregulated in ginger water-treated groups. These results clearly revealed the lowering body weight gain effect of ginger water, which most likely occurs at the transcriptional level of energy metabolizing proteins.
Collapse
Affiliation(s)
- Samy Sayed
- Department of Biotechnology, Faculty of Science, Taif University, Taif 21974, Saudi Arabia; (S.S.); (A.E.-S.); (M.A.); (S.A.-O.); (S.F.)
- Faculty of Agriculture, Cairo University, Giza 12613, Egypt
| | - Mohamed Ahmed
- Faculty of Veterinary Medicine, University of Sadat City, Sadat City 32958, Egypt;
| | - Ahmed El-Shehawi
- Department of Biotechnology, Faculty of Science, Taif University, Taif 21974, Saudi Arabia; (S.S.); (A.E.-S.); (M.A.); (S.A.-O.); (S.F.)
- Department of Genetics, Faculty of Agriculture, University of Alexandria, Alexandria 21526, Egypt
| | - Mohamed Alkafafy
- Department of Biotechnology, Faculty of Science, Taif University, Taif 21974, Saudi Arabia; (S.S.); (A.E.-S.); (M.A.); (S.A.-O.); (S.F.)
- Faculty of Veterinary Medicine, University of Sadat City, Sadat City 32958, Egypt;
| | - Saqer Al-Otaibi
- Department of Biotechnology, Faculty of Science, Taif University, Taif 21974, Saudi Arabia; (S.S.); (A.E.-S.); (M.A.); (S.A.-O.); (S.F.)
| | - Hanan El-Sawy
- Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt;
| | - Samy Farouk
- Department of Biotechnology, Faculty of Science, Taif University, Taif 21974, Saudi Arabia; (S.S.); (A.E.-S.); (M.A.); (S.A.-O.); (S.F.)
| | - Samir El-Shazly
- Department of Biotechnology, Faculty of Science, Taif University, Taif 21974, Saudi Arabia; (S.S.); (A.E.-S.); (M.A.); (S.A.-O.); (S.F.)
- Department of Biochemistry, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr Elsheikh 33511, Egypt
| |
Collapse
|
37
|
Asadi P, Vessal M, Khorsand M, Takhshid MA. Erythrocyte glucose-6-phosphate dehydrogenase activity and risk of gestational diabetes. J Diabetes Metab Disord 2020; 18:533-541. [PMID: 31890679 DOI: 10.1007/s40200-019-00464-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/31/2019] [Indexed: 12/29/2022]
Abstract
Purpose Glucose-6-phosphate dehydrogenase (G6PD) is the regulating enzyme in the pentose phosphate pathway. A link between the activity of G6PD and diabetes mellitus has previously been reported. The association of G6PD activity with the pathogenesis of gestational diabetes mellitus (GDM) has not yet been investigated. The aim of the present study was to investigate the association of erythrocyte G6PD activity with major characteristics of GDM. Methods This case-control study was conducted at Hafez Hospital, Shiraz University of Medical Sciences, Shiraz, Iran from March to November 2017. Eighty-four age-matched pregnant women including GDM (n = 33), impaired glucose tolerance (IGT; n = 7), and normal glucose tolerance (NGT; n = 44) subjects were enrolled in this study. The levels of erythrocyte G6PD activity, fasting plasma glucose (FPG), insulin, malondialdehyde (MDA), and ferric reducing power (FRAP) of serum were measured. The level of homeostasis model for the assessment of insulin resistance (HOMA-IR) was calculated. The data were analyzed using SPSS software. P < 0.05 was considered statistically significant. Results The values of FPG, insulin, HOMA-IR, G6PD activity, and FRAP were significantly higher in GDM patients compared to NGT subjects. G6PD activity was correlated with FPG ((r = 0.224; P = 0.041). Binary logistic regression analysis revealed independent association of body mass index >25.88 [OR = 3.23, 95% CI 1.071-9.75, P = 0.037], HOMA- IR >2.33 [OR = 7.15, 95% CI 2.26-22.56, P < 0.001], and G6PD activity>21.17 U/g Hb [OR = 4.63, 95% CI 1.49-14.38, P = 0.008] with an increased risk of GDM. No significant change was observed among serum MDA levels in the three groups. Conclusion The findings demonstrate that increased G6PD activity is positively associated with the risk of GDM.
Collapse
Affiliation(s)
- Parvaneh Asadi
- 1Department of Molecular Biology-Biochemistry, Islamic Azad University, Shiraz Branch, Shiraz, Iran
| | - Mahmood Vessal
- 1Department of Molecular Biology-Biochemistry, Islamic Azad University, Shiraz Branch, Shiraz, Iran
| | - Marjan Khorsand
- 2Diagnostic Laboratory Sciences and Technology Research Center, Faculty of Paramedical Sciences, Shiraz University of Medical Sciences, Meshkinfam Street, Shiraz, Iran
| | - Mohammad Ali Takhshid
- 2Diagnostic Laboratory Sciences and Technology Research Center, Faculty of Paramedical Sciences, Shiraz University of Medical Sciences, Meshkinfam Street, Shiraz, Iran
| |
Collapse
|
38
|
Tu D, Gao Y, Yang R, Guan T, Hong JS, Gao HM. The pentose phosphate pathway regulates chronic neuroinflammation and dopaminergic neurodegeneration. J Neuroinflammation 2019; 16:255. [PMID: 31805953 PMCID: PMC6896486 DOI: 10.1186/s12974-019-1659-1] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 11/26/2019] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Metabolic dysfunction and neuroinflammation are increasingly implicated in Parkinson's disease (PD). The pentose phosphate pathway (PPP, a metabolic pathway parallel to glycolysis) converts glucose-6-phosphate into pentoses and generates ribose-5-phosphate and NADPH thereby governing anabolic biosynthesis and redox homeostasis. Brains and immune cells display high activity of glucose-6-phosphate dehydrogenase (G6PD), the rate-limiting enzyme of the PPP. A postmortem study reveals dysregulation of G6PD enzyme in brains of PD patients. However, spatial and temporal changes in activity/expression of G6PD in PD remain undetermined. More importantly, it is unclear how dysfunction of G6PD and the PPP affects neuroinflammation and neurodegeneration in PD. METHODS We examined expression/activity of G6PD and its association with microglial activation and dopaminergic neurodegeneration in multiple chronic PD models generated by an intranigral/intraperitoneal injection of LPS, daily subcutaneous injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) for 6 days, or transgenic expression of A53T α-synuclein. Primary microglia were transfected with G6PD siRNAs and treated with lipopolysaccharide (LPS) to examine effects of G6PD knockdown on microglial activation and death of co-cultured neurons. LPS alone or with G6PD inhibitor(s) was administrated to mouse substantia nigra or midbrain neuron-glia cultures. While histological and biochemical analyses were conducted to examine microglial activation and dopaminergic neurodegeneration in vitro and in vivo, rotarod behavior test was performed to evaluate locomotor impairment in mice. RESULTS Expression and activity of G6PD were elevated in LPS-treated midbrain neuron-glia cultures (an in vitro PD model) and the substantia nigra of four in vivo PD models. Such elevation was positively associated with microglial activation and dopaminergic neurodegeneration. Furthermore, inhibition of G6PD by 6-aminonicotinamide and dehydroepiandrosterone and knockdown of microglial G6PD attenuated LPS-elicited chronic dopaminergic neurodegeneration. Mechanistically, microglia with elevated G6PD activity/expression produced excessive NADPH and provided abundant substrate to over-activated NADPH oxidase (NOX2) leading to production of excessive reactive oxygen species (ROS). Knockdown and inhibition of G6PD ameliorated LPS-triggered production of ROS and activation of NF-кB thereby dampening microglial activation. CONCLUSIONS Our findings indicated that G6PD-mediated PPP dysfunction and neuroinflammation exacerbated each other mediating chronic dopaminergic neurodegeneration and locomotor impairment. Insight into metabolic-inflammatory interface suggests that G6PD and NOX2 are potential therapeutic targets for PD.
Collapse
Affiliation(s)
- Dezhen Tu
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, 12 Xuefu Road, Nanjing, 210061, Jiangsu Province, China
- Neurobiology Laboratory, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, Durham, NC, 27709, USA
| | - Yun Gao
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, 12 Xuefu Road, Nanjing, 210061, Jiangsu Province, China
- Neurobiology Laboratory, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, Durham, NC, 27709, USA
| | - Ru Yang
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, 12 Xuefu Road, Nanjing, 210061, Jiangsu Province, China
| | - Tian Guan
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, 12 Xuefu Road, Nanjing, 210061, Jiangsu Province, China
| | - Jau-Shyong Hong
- Neurobiology Laboratory, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, Durham, NC, 27709, USA
| | - Hui-Ming Gao
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, 12 Xuefu Road, Nanjing, 210061, Jiangsu Province, China.
- Neurobiology Laboratory, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, Durham, NC, 27709, USA.
| |
Collapse
|
39
|
Jiang A, Dong C, Li B, Zhang Z, Chen Y, Ning C, Wu W, Liu H. MicroRNA-206 regulates cell proliferation by targeting G6PD in skeletal muscle. FASEB J 2019; 33:14083-14094. [PMID: 31675481 DOI: 10.1096/fj.201900502rrrr] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Skeletal muscle is a major component of body mass and plays a central role in the control of whole-body metabolism in humans and animals. Therefore, elucidation of the underlying mechanisms of skeletal growth and development are expected to lead to the discovery of novel genes and pathways related to muscle disease. miR-206, a skeletal muscle-specific microRNA, plays a crucial role in myogenesis; however, miR-206 is known to function in myogenic differentiation, whether or not it affects muscle cells' proliferation, and the underlying mechanisms are unknown. In this study, we investigated the effect of miR-206 on muscle cell proliferation and differentiation, as well as its effect on myofiber type conversion using mouse C2C12 myoblasts. The results showed that overexpression of miR-206 inhibited cell proliferation and promoted muscle cell differentiation, but it did not affect myofiber type conversion. Intriguingly, we found that overexpression of miR-206 suppressed muscle cell proliferation and induced cell cycle arrest in G0/G1 phase by inhibiting the glucose-6-phosphate dehydrogenase (G6PD) gene. Taken together, we demonstrated that the miR-206-G6PD pathway suppresses muscle cell proliferation, and these findings may facilitate the treatment of muscle diseases.-Jiang, A., Dong, C., Li, B., Zhang, Z., Chen, Y., Ning, C., Wu, W., Liu, H. MicroRNA-206 regulates cell proliferation by targeting G6PD in skeletal muscle.
Collapse
Affiliation(s)
- Aiwen Jiang
- Department of Animal Genetics, Breeding, and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Chao Dong
- Department of Animal Genetics, Breeding, and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Bojiang Li
- Department of Animal Genetics, Breeding, and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.,College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Zengkai Zhang
- Department of Animal Genetics, Breeding, and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yujun Chen
- Department of Animal Genetics, Breeding, and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Caibo Ning
- Department of Animal Genetics, Breeding, and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Wangjun Wu
- Department of Animal Genetics, Breeding, and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Honglin Liu
- Department of Animal Genetics, Breeding, and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
40
|
Xue J, Chen TT, Zheng JW, Balamurugan S, Liu YH, Yang WD, Liu JS, Li HY. Glucose-6-Phosphate Dehydrogenase from the Oleaginous Microalga Nannochloropsis Uncovers Its Potential Role in Promoting Lipogenesis. Biotechnol J 2019; 15:e1900135. [PMID: 31464064 DOI: 10.1002/biot.201900135] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 08/20/2019] [Indexed: 12/15/2022]
Abstract
Microalgae have long been considered as potential biological feedstock for the production of wide array of bioproducts, such as biofuel feedstock because of their lipid accumulating capability. However, lipid productivity of microalgae is still far below commercial viability. Here, a glucose-6-phosphate dehydrogenase from the oleaginous microalga Nannochloropsis oceanica is identified and heterologously expressed in the green microalga Chlorella pyrenoidosa to characterize its function in the pentose phosphate pathway. It is found that the G6PD enzyme activity toward NADPH production is increased by 2.19-fold in engineered microalgal strains. Lipidomic analysis reveals up to 3.09-fold increase of neutral lipid content in the engineered strains, and lipid yield is gradually increased throughout the cultivation phase and saturated at the stationary phase. Moreover, cellular physiological characteristics including photosynthesis and growth rate are not impaired. Collectively, these results reveal the pivotal role of glucose-6-phosphate dehydrogenase from N. oceanica in NADPH supply, demonstrating that provision of reducing power is crucial for microalgal lipogenesis and can be a potential target for metabolic engineering.
Collapse
Affiliation(s)
- Jiao Xue
- Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.,The College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Ting-Ting Chen
- Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Jian-Wei Zheng
- Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Srinivasan Balamurugan
- Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Yu-Hong Liu
- Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Wei-Dong Yang
- Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Jie-Sheng Liu
- Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Hong-Ye Li
- Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| |
Collapse
|
41
|
Maciukiewicz M, Tiwari AK, Zai CC, Gorbovskaya I, Laughlin CP, Nurmi EL, Liebermann JA, Meltzer HY, Kennedy JL, Müller DJ. Genome-wide association study on antipsychotic-induced weight gain in Europeans and African-Americans. Schizophr Res 2019; 212:204-212. [PMID: 31447353 DOI: 10.1016/j.schres.2019.07.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 06/13/2019] [Accepted: 07/11/2019] [Indexed: 01/07/2023]
Abstract
BACKGROUND Antipsychotic (AP) medications are the first line of treatment for schizophrenia. However, most conferr a risk of antipsychotic-induced weight gain (AIWG). The objective of this investigation was to conduct a genome-wide association study (GWAS) of AIWG, followed by comprehensive, post-GWAS approaches. METHODS We investigated n = 201 schizophrenia or schizoaffective disorder patients of European and African American ancestry who were treated primarily with clozapine or olanzapine. We conducted a genome-wide association analysis for AIWG, defined primarily as a percentage of weight change from baseline. RESULTS When examining Europeans (n = 147), we noticed an association between rs62097526 (β = 0.39, p = 3.59 × 10-6, CADD = 2.213) variant, located downstream of the CIDEA gene, which is considered a risk factor for AIWG. In the entire sample, we observed a significant association between rs1525085 (β = 0.411, p = 3.15 × 10-9) variant of the DGKB gene and AIWG. The association was nominally significant in Europeans (β = 0.271, p = 0.002) and African Americans (β = 0.579, p = 5.73 × 10-5) with the same risk allele. Our top genes (p < 5 × 10-5) were enriched in the GWAS catalog for the risk of obesity and interacted with the known risk factors for obesity (G6PD) and diabetes (IRS1). In addition, these genes are targeted by miRNAs related to schizophrenia (mir-34a) and obesity (mir-19b). However, our polygenic risk score analyses did not provide support for major genetic overlap between obesity and the risk of AIWG. CONCLUSIONS In summary, we propose that the CIDEA and DGKB genes are risk factors for AIWG in transethnic populations. Additionally, our evidence suggests that the G6PD and IRS1 gene-related pathways might be involved in AIWG.
Collapse
Affiliation(s)
- Malgorzata Maciukiewicz
- Pharmacogenetic Research Clinic, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Arun K Tiwari
- Pharmacogenetic Research Clinic, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Clement C Zai
- Pharmacogenetic Research Clinic, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Ilona Gorbovskaya
- Pharmacogenetic Research Clinic, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Christopher P Laughlin
- Division of Child and Adolescent Psychiatry, UCLA Semel Institute for Neuroscience, Los Angeles, California, USA
| | - Erika L Nurmi
- Division of Child and Adolescent Psychiatry, UCLA Semel Institute for Neuroscience, Los Angeles, California, USA
| | - Jeffrey A Liebermann
- Department of Psychiatry, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Herbert Y Meltzer
- Department of Psychiatry and Behavioral Sciences, Northwestern University, Chicago, IL, USA
| | - James L Kennedy
- Pharmacogenetic Research Clinic, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Daniel J Müller
- Pharmacogenetic Research Clinic, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
42
|
Jang M, Choi HY, Kim GH. Phenolic components rich ethyl acetate fraction of Orostachys japonicus inhibits lipid accumulation by regulating reactive oxygen species generation in adipogenesis. J Food Biochem 2019; 43:e12939. [PMID: 31368568 DOI: 10.1111/jfbc.12939] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 03/13/2019] [Accepted: 04/23/2019] [Indexed: 12/11/2022]
Abstract
In this study, Orostachys japonicus was extracted with ethyl alcohol and fractionated by a serial of organic solvents. The ethyl acetate fraction was found to be the most effective among the tested five fractions. High-performance liquid chromatography and mass spectrometry analysis of the ethyl acetate fraction presented epicatechin gallate, quercetin-3-O-glucoside, and kaempferol-3-O-rutinoside. Treatment with O. japonicus inhibited reactive oxygen species (ROS) generation and lipid accumulation during adipogenesis. The gene expression of enzymes involved in the antioxidant system increased in O. japonicus-treated cells. messeanger RNA (mRNA) and protein expression of the pro-oxidant enzymes such as nicotinamide adenine dinucleotide phosphate hydrogen oxidase4 and glucose-6-phosphate dehydrogenase suppressed in O. japonicus-treated cells. O. japonicus also inhibited the mRNA and protein levels of adipogenic transcription factors (including proliferator activated receptor-γ and CCAAT/enhancer-binding protein-α) and their target gene (adipocyte protein 2). These results suggest that O. japonicus inhibits adipogenesis by controlling pro-/anti-oxidant enzyme responses and adipogenic transcription factors. PRACTICAL APPLICATIONS: ROS generation is markedly related to the pathogenesis and development of metabolic disorders. Treatment with O. japonicus inhibited ROS generation and lipid accumulation during adipogenesis. This result indicates that O. japonicus inhibit adipogenesis by controlling pro-/anti-oxidant enzyme responses and adipogenic mediators.
Collapse
Affiliation(s)
- Miran Jang
- Plant Resources Research Institute, Duksung Women's University, Seoul, Republic of Korea
| | - Hye-Young Choi
- Plant Resources Research Institute, Duksung Women's University, Seoul, Republic of Korea
| | - Gun-Hee Kim
- Plant Resources Research Institute, Duksung Women's University, Seoul, Republic of Korea
| |
Collapse
|
43
|
Park SB, Lee SY, Jung WH, Lee J, Jeong HG, Hong J, Kang D, Kim KY. Development of in vitro three-dimensional co-culture system for metabolic syndrome therapeutic agents. Diabetes Obes Metab 2019; 21:1146-1157. [PMID: 30609258 DOI: 10.1111/dom.13628] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 12/23/2018] [Accepted: 01/02/2019] [Indexed: 01/08/2023]
Abstract
AIMS There are many obstacles to overcome in the development of new drugs for metabolic diseases, including efficacy and toxicity problems in later stages of drug development. To overcome these problems and predict efficacy and toxicity in early stages, we constructed a new model of insulin resistance in terms of communication between 3T3-L1 adipocytes and RAW264.7 macrophages by three-dimensional (3D) culture. RESULTS In this study, results focused on the functional resemblance between 3D co-culture of adipocytes and macrophages and adipose tissue in diabetic mice. The 3D mono-culture preadipocytes showed good cell viability and induced cell differentiation to adipocytes, without cell confluence or cell-cell contact and interaction. The 3D co-cultured preadipocytes with RAW264.7 macrophages induced greater insulin resistance than two-dimensional and 3D mono-cultured adipocytes. Additionally, we demonstrated that 3D co-culture model had functional metabolic similarity to adipose tissue in diabetic mice. We utilized this 3D co-culture system to screen PPARγ antagonists that might have potential as therapeutic agents for diabetes as demonstrated by an in vivo assay. CONCLUSION This in vitro 3D co-culture system could serve as a next-generation platform to accelerate the development of therapeutics for metabolic diseases.
Collapse
Affiliation(s)
- Sung B Park
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Sun Y Lee
- College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| | - Won H Jung
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Junhee Lee
- Department of Nature-Inspired Nano Convergence System, Korea Institute of Machinery and Materials, Daejeon, Republic of Korea
| | - Hye G Jeong
- Department of Toxicology, College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Jongki Hong
- College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| | - Dukjin Kang
- Center for Bioanalysis, Division of Chemical and Medical Metrology, Korea Research Institute of Standards and Science, Daejeon, Republic of Korea
| | - Ki Y Kim
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
44
|
Wang M, Hu J, Yan L, Yang Y, He M, Wu M, Li Q, Gong W, Yang Y, Wang Y, Handy DE, Lu B, Hao C, Wang Q, Li Y, Hu R, Stanton RC, Zhang Z. High glucose-induced ubiquitination of G6PD leads to the injury of podocytes. FASEB J 2019; 33:6296-6310. [PMID: 30785802 DOI: 10.1096/fj.201801921r] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Oxidative stress contributes substantially to podocyte injury, which plays an important role in the development of diabetic kidney disease. The mechanism of hyperglycemia-induced oxidative stress in podocytes is not fully understood. Glucose-6-phosphate dehydrogenase (G6PD) is critical in maintaining NADPH, which is an important cofactor for the antioxidant system. Here, we hypothesized that high glucose induced ubiquitination and degradation of G6PD, which injured podocytes by reactive oxygen species (ROS) accumulation. We found that G6PD protein expression was decreased in kidneys of both diabetic patients and diabetic rodents. G6PD activity was also reduced in diabetic mice. Overexpressing G6PD reversed redox imbalance and podocyte apoptosis induced by high glucose and palmitate. Inhibition of G6PD with small interfering RNA induced podocyte apoptosis. In kidneys of G6PD-deficient mice, podocyte apoptosis was significantly increased. Interestingly, high glucose had no effect on G6PD mRNA expression. Decreased G6PD protein expression was mediated by the ubiquitin proteasome pathway. We found that the von Hippel-Lindau (VHL) protein, an E3 ubiquitin ligase subunit, directly bound to G6PD and degraded G6PD through ubiquitylating G6PD on K366 and K403. In summary, our data suggest that high glucose induces ubiquitination of G6PD by VHL E3 ubiquitin ligase, which leads to ROS accumulation and podocyte injury.-Wang, M., Hu, J., Yan, L., Yang, Y., He, M., Wu, M., Li, Q., Gong, W., Yang, Y., Wang, Y., Handy, D. E., Lu, B., Hao, C., Wang, Q., Li, Y., Hu, R., Stanton, R. C., Zhang, Z. High glucose-induced ubiquitination of G6PD leads to the injury of podocytes.
Collapse
Affiliation(s)
- Meng Wang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Ji Hu
- Department of Endocrinology, The Second Affiliated Hospital, Soochow University, Suzhou, China
| | - Linling Yan
- Department of Endocrinology, The First People's Hospital of Taicang, Suzhou, China
| | - Yeping Yang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Min He
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Meng Wu
- Department of Endocrinology, The Second Affiliated Hospital, Soochow University, Suzhou, China
| | - Qin Li
- Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Wei Gong
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Yang Yang
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Shanghai, China
| | - Yi Wang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Diane E Handy
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Bin Lu
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Chuanming Hao
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qinghua Wang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China.,Division of Endocrinology and Metabolism, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Yiming Li
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Ronggui Hu
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Shanghai, China
| | - Robert C Stanton
- Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Zhaoyun Zhang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
45
|
Abstract
Adipose tissue possesses the remarkable capacity to control its size and function in response to a variety of internal and external cues, such as nutritional status and temperature. The regulatory circuits of fuel storage and oxidation in white adipocytes and thermogenic adipocytes (brown and beige adipocytes) play a central role in systemic energy homeostasis, whereas dysregulation of the pathways is closely associated with metabolic disorders and adipose tissue malfunction, including obesity, insulin resistance, chronic inflammation, mitochondrial dysfunction, and fibrosis. Recent studies have uncovered new regulatory elements that control the above parameters and provide new mechanistic opportunities to reprogram fat cell fate and function. In this Review, we provide an overview of the current understanding of adipocyte metabolism in physiology and disease and also discuss possible strategies to alter fuel utilization in fat cells to improve metabolic health.
Collapse
Affiliation(s)
- Edward T Chouchani
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
| | - Shingo Kajimura
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA.
- UCSF Diabetes Center, San Francisco, CA, USA.
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA, USA.
| |
Collapse
|
46
|
Kumar N, Singh AK. Reactive oxygen species in seminal plasma as a cause of male infertility. J Gynecol Obstet Hum Reprod 2018; 47:565-572. [DOI: 10.1016/j.jogoh.2018.06.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 06/13/2018] [Accepted: 06/21/2018] [Indexed: 12/31/2022]
|
47
|
Liu Y, Zheng J, Zhang HP, Zhang X, Wang L, Wood L, Wang G. Obesity-Associated Metabolic Signatures Correlate to Clinical and Inflammatory Profiles of Asthma: A Pilot Study. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2018; 10:628-647. [PMID: 30306746 PMCID: PMC6182193 DOI: 10.4168/aair.2018.10.6.628] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/27/2018] [Accepted: 06/01/2018] [Indexed: 02/05/2023]
Abstract
PURPOSE Obesity is associated with metabolic dysregulation, but the underlying metabolic signatures involving clinical and inflammatory profiles of obese asthma are largely unexplored. We aimed at identifying the metabolic signatures of obese asthma. METHODS Eligible subjects with obese (n = 11) and lean (n = 22) asthma underwent body composition and clinical assessment, sputum induction, and blood sampling. Sputum supernatant was assessed for interleukin (IL)-1β, -4, -5, -6, -13, and tumor necrosis factor (TNF)-α, and serum was detected for leptin, adiponectin and C-reactive protein. Untargeted gas chromatography time-of-flight mass spectrometry (GC-TOF-MS)-based metabolic profiles in sputum, serum and peripheral blood monocular cells (PBMCs) were analyzed by orthogonal projections to latent structures-discriminate analysis (OPLS-DA) and pathway topology enrichment analysis. The differential metabolites were further validated by correlation analysis with body composition, and clinical and inflammatory profiles. RESULTS Body composition, asthma control, and the levels of IL-1β, -4, -13, leptin and adiponectin in obese asthmatics were significantly different from those in lean asthmatics. OPLS-DA analysis revealed 28 differential metabolites that distinguished obese from lean asthmatic subjects. The validation analysis identified 18 potential metabolic signatures (11 in sputum, 4 in serum and 2 in PBMCs) of obese asthmatics. Pathway topology enrichment analysis revealed that cyanoamino acid metabolism, caffeine metabolism, alanine, aspartate and glutamate metabolism, phenylalanine, tyrosine and tryptophan biosynthesis, pentose phosphate pathway in sputum, and glyoxylate and dicarboxylate metabolism, glycerolipid metabolism and pentose phosphate pathway in serum are suggested to be significant pathways related to obese asthma. CONCLUSIONS GC-TOF-MS-based metabolomics indicates obese asthma is characterized by a metabolic profile different from lean asthma. The potential metabolic signatures indicated novel immune-metabolic mechanisms in obese asthma with providing more phenotypic and therapeutic implications, which needs further replication and validation.
Collapse
Affiliation(s)
- Ying Liu
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Zheng
- Department of Integrated Traditional Chinese and Western Medicine, Xinqiao Hospital, Third Military University, Chongqing, China
| | - Hong Ping Zhang
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Zhang
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Lei Wang
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Lisa Wood
- Center for Asthma and Respiratory Diseases, Department of Respiratory and Sleep Medicine, John Hunter Hospital, Hunter Medical Research Institute, University of Newcastle, New Lambton, NSW, Australia
| | - Gang Wang
- Department of Respiratory and Critical Care Medicine, Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
48
|
Xue J, Chen TT, Zheng JW, Balamurugan S, Cai JX, Liu YH, Yang WD, Liu JS, Li HY. The role of diatom glucose-6-phosphate dehydrogenase on lipogenic NADPH supply in green microalgae through plastidial oxidative pentose phosphate pathway. Appl Microbiol Biotechnol 2018; 102:10803-10815. [PMID: 30349933 DOI: 10.1007/s00253-018-9415-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 08/23/2018] [Accepted: 09/20/2018] [Indexed: 11/27/2022]
Abstract
Commercial production of biofuel from oleaginous microalgae is often impeded by their slow growth rate than other fast-growing algal species. A promising strategy is to genetically engineer the fast-growing algae to accumulate lipids by expressing key lipogenic genes from oleaginous microalgae. However, lacking of strong expression cassette to transform most of the algal species and potential metabolic target to engineer lipid metabolism has hindered its biotechnological applications. In this study, we engineered the oxidative pentose phosphate pathway (PPP) of green microalga Chlorella pyrenoidosa for lipid enhancement by expressing a glucose-6-phosphate dehydrogenase (G6PD) from oleaginous diatom Phaeodactylum tricornutum. Molecular characterization of transformed lines revealed that heterologous PtG6PD was transcribed and expressed successfully. Interestingly, subcellular localization analyses revealed that PtG6PD was targeted to chloroplasts of C. pyrenoidosa. PtG6PD expression remarkably elevated NADPH content and consequently enhanced the lipid content without affecting growth rate. Collectively, this report represents a promising candidate to engineer lipid biosynthesis in heterologous hosts with notable commercial significance, and it highlights the potential role of plastidial PPP in supplying lipogenic NADPH in microalgae.
Collapse
Affiliation(s)
- Jiao Xue
- Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Ting-Ting Chen
- Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Jian-Wei Zheng
- Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Srinivasan Balamurugan
- Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Jia-Xi Cai
- Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Yu-Hong Liu
- Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Wei-Dong Yang
- Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Jie-Sheng Liu
- Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Hong-Ye Li
- Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
49
|
Inhibitory effects of Orostachys malacophyllus var. iwarenge extracts on reactive oxygen species production and lipid accumulation during 3T3-L1 adipocyte differentiation. Food Sci Biotechnol 2018; 28:227-236. [PMID: 30815314 DOI: 10.1007/s10068-018-0426-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 06/19/2018] [Accepted: 07/01/2018] [Indexed: 01/08/2023] Open
Abstract
Orostachys malacophyllus var. iwarenge was extracted with water, 30 and 70% ethyl alcohol. The ethyl alcohol extracts showed higher total phenol contents and greater antioxidant effects than the water extract. Treatment with the 70% alcohol extract inhibited reactive oxygen species (ROS) production and lipid accumulation during 3T3-L1 adipogenesis. Furthermore, the 70% extract inhibited the mRNA and protein expressions of the pro-oxidant enzyme NADPH oxidase 4 and of the NADPH-producing enzyme glucose-6-phosphate dehydrogenase. The mRNA and protein expressions of antioxidant enzymes, such as copper-zinc superoxide dismutase and manganese superoxide dismutase increased in cells treated with the 70% alcohol extract. In addition, this extract suppressed the mRNA and protein levels of adipogenic transcription factors and of their marker genes. These results indicate that O. malacophyllus extracts inhibit lipid accumulation and ROS production by controlling adipogenic factors and pro-/anti-oxidant enzyme responses.
Collapse
|
50
|
Lemecha M, Morino K, Seifu D, Imamura T, Nakagawa F, Nagata A, Okamato T, Sekine O, Ugi S, Maegawa H. Improved glucose metabolism by Eragrostis tef potentially through beige adipocyte formation and attenuating adipose tissue inflammation. PLoS One 2018; 13:e0201661. [PMID: 30071087 PMCID: PMC6072038 DOI: 10.1371/journal.pone.0201661] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 07/19/2018] [Indexed: 12/31/2022] Open
Abstract
Background Teff is a staple food in Ethiopia that is rich in dietary fiber. Although gaining popularity in Western countries because it is gluten-free, the effects of teff on glucose metabolism remain unknown. Aim To evaluate the effects of teff on body weight and glucose metabolism compared with an isocaloric diet containing wheat. Results Mice fed teff weighed approximately 13% less than mice fed wheat (p < 0.05). The teff-based diet improved glucose tolerance compared with the wheat group with normal chow but not with a high-fat diet. Reduced adipose inflammation characterized by lower expression of TNFα, Mcp1, and CD11c, together with higher levels of cecal short chain fatty acids such as acetate, compared with the control diet containing wheat after 14 weeks of dietary treatment. In addition, beige adipocyte formation, characterized by increased expression of Ucp-1 (~7-fold) and Cidea (~3-fold), was observed in the teff groups compared with the wheat group. Moreover, a body-weight matched experiment revealed that teff improved glucose tolerance in a manner independent of body weight reduction after 6 weeks of dietary treatment. Enhanced beige adipocyte formation without improved adipose inflammation in a body-weight matched experiment suggests that the improved glucose metabolism was a consequence of beige adipocyte formation, but not solely through adipose inflammation. However, these differences between teff- and wheat-containing diets were not observed in the high-fat diet group. Conclusions Teff improved glucose tolerance likely by promoting beige adipocyte formation and improved adipose inflammation.
Collapse
Affiliation(s)
- Mengistu Lemecha
- Division of Endocrinology and Metabolism, Department of Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Katsutaro Morino
- Division of Endocrinology and Metabolism, Department of Medicine, Shiga University of Medical Science, Otsu, Japan
- * E-mail:
| | - Daniel Seifu
- School of Medicine, Department of Biochemistry, Addis Ababa University, Addis Ababa, Ethiopia
| | - Takeshi Imamura
- Division of Molecular Pharmacology, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Fumiyuki Nakagawa
- Division of Endocrinology and Metabolism, Department of Medicine, Shiga University of Medical Science, Otsu, Japan
- CMIC Pharma Science, Osaka, Japan
| | | | - Takuya Okamato
- Division of Endocrinology and Metabolism, Department of Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Osamu Sekine
- Division of Endocrinology and Metabolism, Department of Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Satoshi Ugi
- Division of Endocrinology and Metabolism, Department of Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Hiroshi Maegawa
- Division of Endocrinology and Metabolism, Department of Medicine, Shiga University of Medical Science, Otsu, Japan
| |
Collapse
|