1
|
Tong W, Allison BJ, Brain KL, Patey OV, Niu Y, Botting KJ, Ford SG, Garrud TA, Wooding PFB, Lyu Q, Zhang L, Ma J, Sowton AP, O'Brien KA, Cindrova-Davies T, Yung HW, Burton GJ, Murray AJ, Giussani DA. Placental mitochondrial metabolic adaptation maintains cellular energy balance in pregnancy complicated by gestational hypoxia. J Physiol 2025. [PMID: 39868991 DOI: 10.1113/jp287897] [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: 10/17/2024] [Accepted: 12/18/2024] [Indexed: 01/28/2025] Open
Abstract
The mechanisms that drive placental dysfunction in pregnancies complicated by hypoxia and fetal growth restriction remain poorly understood. Changes to mitochondrial respiration contribute to cellular dysfunction in conditions of hypoxia and have been implicated in the pathoaetiology of pregnancy complications, such as pre-eclampsia. We used bespoke isobaric hypoxic chambers and a combination of functional, molecular and imaging techniques to study cellular metabolism and mitochondrial dynamics in sheep undergoing hypoxic pregnancy. We show that hypoxic pregnancy in sheep triggers a shift in capacity away from β-oxidation and complex I-mediated respiration, while maintaining total oxidative phosphorylation capacity. There are also complex-specific changes to electron transport chain composition and a switch in mitochondrial dynamics towards fission. Hypoxic placentas show increased activation of the non-canonical mitochondrial unfolded protein response pathway and enhanced insulin like growth factor 2 signalling. Combined, therefore, the data show that the hypoxic placenta undergoes significant metabolic and morphological adaptations to maintain cellular energy balance. Chronic hypoxia during pregnancy in sheep activated placental mitochondrial stress pathways, leading to alterations in mitochondrial respiration, mitochondrial energy metabolism and mitochondrial dynamics, as seen in the placenta of women with pre-eclampsia. KEY POINTS: Hypoxia shifts mitochondrial respiration away from β-oxidation and complex I. Complex-specific changes occur in the electron transport chain composition. Activation of the non-canonical mitochondrial unfolded protein response pathway is heightened in hypoxic placentas. Enhanced insulin like growth factor 2 signalling is observed in hypoxic placentas. Hypoxic placentas undergo significant functional adaptations for energy balance.
Collapse
Affiliation(s)
- Wen Tong
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
| | - Beth J Allison
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Kirsty L Brain
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Olga V Patey
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Youguo Niu
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
- Cardiovascular Strategic Research Initiative, University of Cambridge, Cambridge, UK
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| | - Kimberley J Botting
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
- Cardiovascular Strategic Research Initiative, University of Cambridge, Cambridge, UK
| | - Sage G Ford
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Tess A Garrud
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Peter F B Wooding
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Qiang Lyu
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| | - Lin Zhang
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| | - Jin Ma
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| | - Alice P Sowton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Katie A O'Brien
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Tereza Cindrova-Davies
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
| | - Hong Wa Yung
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
| | - Graham J Burton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
| | - Andrew J Murray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Cardiovascular Strategic Research Initiative, University of Cambridge, Cambridge, UK
| | - Dino A Giussani
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
- Cardiovascular Strategic Research Initiative, University of Cambridge, Cambridge, UK
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
2
|
Spinedi E, Docena GH. Physiopathological Roles of White Adiposity and Gut Functions in Neuroinflammation. Int J Mol Sci 2024; 25:11741. [PMID: 39519291 PMCID: PMC11546880 DOI: 10.3390/ijms252111741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
White adipose tissue (WAT) and the gut are involved in the development of neuroinflammation when an organism detects any kind of injury, thereby triggering metainflammation. In fact, the autonomous nervous system innervates both tissues, although the complex role played by the integrated sympathetic, parasympathetic, and enteric nervous system functions have not been fully elucidated. Our aims were to investigate the participation of inflamed WAT and the gut in neuroinflammation. Firstly, we conducted an analysis into how inflamed peripheral WAT plays a key role in the triggering of metainflammation. Indeed, this included the impact of the development of local insulin resistance and its metabolic consequences, a serious hypothalamic dysfunction that promotes neurodegeneration. Then, we analyzed the gut-brain axis dysfunction involved in neuroinflammation by examining cell interactions, soluble factors, the sensing of microbes, and the role of dysbiosis-related mechanisms (intestinal microbiota and mucosal barriers) affecting brain functions. Finally, we targeted the physiological crosstalk between cells of the brain-WAT-gut axis that restores normal tissue homeostasis after injury. We concluded the following: because any injury can result not only in overall insulin resistance and dysbiosis, which in turn can impact upon the brain, but that a high-risk of the development of neuroinflammation-induced neurodegenerative disorder can also be triggered. Thus, it is imperative to avoid early metainflammation by applying appropriate preventive (e.g., lifestyle and diet) or pharmacological treatments to cope with allostasis and thus promote health homeostasis.
Collapse
Affiliation(s)
- Eduardo Spinedi
- Centro de Endocrinología Experimental y Aplicada (CENEXA-UNLP-CONICET-CICPBA), University of La Plata Medical School, La Plata 1900, Argentina
| | - Guillermo Horacio Docena
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP-UNLP-CONICET-CICPBA), School of Sciences, University of La Plata, La Plata 1900, Argentina
| |
Collapse
|
3
|
Su D, Song Y, Li D, Yang S, Zhan S, Zhong T, Guo J, Cao J, Li L, Zhang H, Wang L. Cold exposure affects glucose metabolism, lipid droplet deposition and mitophagy in skeletal muscle of newborn goats. Domest Anim Endocrinol 2024; 88:106847. [PMID: 38479188 DOI: 10.1016/j.domaniend.2024.106847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 03/07/2024] [Accepted: 03/07/2024] [Indexed: 06/07/2024]
Abstract
Cold exposure is a common stressor for newborn goats. Skeletal muscle plays an important role in maintaining whole-body homeostasis of glucose and lipid metabolism. However, the molecular mechanisms underlying regulation of skeletal muscle of newborn goats by cold exposure remains unclear. In this study, we found a significant increase (P < 0.01) in serum glucagon levels after 24 h of cold exposure (COLD, 6°C), while glucose and insulin concentrations were significantly decreased (P < 0.01) compared to room temperature (RT, 25°C). Additionally, we found that cold exposure reduced glycogen content (P < 0.01) in skeletal muscle. Pathway enrichment analysis revealed that cold exposure activated skeletal muscle glucose metabolism pathways (including insulin resistance and the insulin signaling pathway) and mitophagy-related pathways. Cold exposure up-regulated the expression of genes involved in fatty acid and triglyceride synthesis, promoting skeletal muscle lipid deposition. Notably, cold exposure induced mitophagy in skeletal muscle.
Collapse
Affiliation(s)
- Duo Su
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yulong Song
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Die Li
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Shizhong Yang
- Institute of Liangshan Agricultural Science Research, Xichang 615042, China
| | - Siyuan Zhan
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Tao Zhong
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiazhong Guo
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiaxue Cao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Li Li
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Hongping Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Linjie Wang
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
4
|
Park SH, Baek SJ, Lee M, Shin HA, Lee HJ, Kim OK, Lee J. Extract mixture of plants (OXYLIA) inhibits fat accumulation by blocking FAS-related factors and promoting lipolysis via cAMP-dependent PKA activation. Food Nutr Res 2024; 68:10180. [PMID: 38571921 PMCID: PMC10989236 DOI: 10.29219/fnr.v68.10180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/14/2023] [Accepted: 12/13/2023] [Indexed: 04/05/2024] Open
Abstract
Background Obesity is characterized by an imbalance between energy intake and expenditure, leading to the excessive accumulation of triglycerides in adipose tissue. Objective This study investigated the potential of Oxylia to prevent obesity in mice fed with a high-fat diet (HFD). Design C57BL/6J mice were fed with one of the following five diets - AIN93G normal diet (normal control), 60% (HFD; control), HFD containing metformin at 40 mg/kg body weight (b.w.) (Met; positive control), HFD containing Oxylia at 30 mg/kg b.w. (O30), or HFD containing Oxylia at 60 mg/kg b.w. (O60) - for 15 weeks. Results Mice under an HFD supplemented with Oxylia had decreased body weight gain, adipose tissue weight, and adipose tissue mass. In addition, triglyceride (TG), total cholesterol, and VLDL/LDL cholesterol levels were lower in the O60 groups than in the HFD-fed control group. Moreover, Oxylia supplementation decreased the expression of adipogenesis-related mRNAs and lipogenesis-related proteins while increasing the expression of lipolysis-related proteins in white adipose tissue and thermogenesis-related proteins in brown adipose tissue. Conclusions These findings suggest that Oxylia has potential as a functional food ingredient for the prevention and treatment of obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Seong-Hoo Park
- Department of Medical Nutrition, Kyung Hee University, Yongin, Republic of Korea
| | - Sun-Jung Baek
- Department of Medical Nutrition, Kyung Hee University, Yongin, Republic of Korea
| | - Minhee Lee
- Department of Medical Nutrition, Kyung Hee University, Yongin, Republic of Korea
| | | | | | - Ok-Kyung Kim
- Division of Food and Nutrition and Human Ecology Research Institute, Chonnam National University, Gwangju, Republic of Korea
| | - Jeongmin Lee
- Department of Medical Nutrition, Kyung Hee University, Yongin, Republic of Korea
| |
Collapse
|
5
|
González-Ibáñez L, Meneses ME, Sánchez-Tapia M, Pérez-Luna D, Torres N, Torre-Villalvazo I, Bonilla M, Petlacalco B, Castillo I, López-Barradas A, Macías A, Tovar AR, Martínez-Carrera D. Edible and medicinal mushrooms ( Pleurotus ostreatus, Ustilago maydis, Ganoderma lucidum) reduce endoplasmic reticulum stress and inflammation in adipose tissue of obese Wistar rats fed with a high fat plus saccharose diet. Food Funct 2023. [PMID: 37161495 DOI: 10.1039/d3fo00089c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Obesity is an increasing global public health problem. A strategy to treat obesity is the use of functional foods. Edible and medicinal mushrooms contain diverse bioactive compounds showing important antihyperlipidemic, antioxidant, and prebiotic properties. We analysed the effects of adding (10%) of Pleurotus ostreatus (Po, basidiomata), Ganoderma lucidum (Gl, basidiomata), or Ustilago maydis (Um, galls), milled, to a high fat plus saccharose diet (HFD + S) for 6 months in a model of obesity with Wistar rats. We assessed weight gain, body composition, lipid parameters, endoplasmic reticulum stress (proteins and inflammatory markers: BiP, XBP-1, JNK, p-JNK, TNF-α), and adiponectin in subcutaneous adipose tissue (SAT). The consumption of edible and medicinal mushrooms decreased weight gain (-17.2-30.1%) and fat mass (-23.7-43.1%), maintained fat-free mass, reduced levels of serum biochemical parameters (TC: -40.1-44.1%, TG: -37.7-51.6%, LDL-C: -64.5-71.1%), and prevented adipocyte hypertrophy (-30.9-36.9%) and collagen deposition (-70.9-73.7%) in SAT. Compared with the HFD + S group, mushroom consumption by Wistar rats significantly reduced the expression of proteins associated with endoplasmic reticulum stress and inflammation (BiP: -72.2-88.2%; XBP-1: -71.5-81.8%; JNK: -71.2-90.0%; p-JNK: -37.3-81.0%; TNF-α: -80.7-91.5%), whereas significantly increased adiponectin protein expression (246.4-654.2%) in SAT. These effects outperformed those obtained through the commercial lipid-lowering drug atorvastatin, contributing synergistically to prevent further obesity-related dysfunctions, such as insulin resistance derived from inflammation and ER stress in adipose tissue. Bioactive compounds from edible, functional and medicinal mushrooms represent new emerging therapies for obesity treatments using natural products.
Collapse
Affiliation(s)
- Laura González-Ibáñez
- Centro de Biotecnología de Hongos Comestibles, Funcionales y Medicinales (CB-HCFM), Campus Puebla, Colegio de Postgraduados (CP), Boulevard Forjadores de Puebla no. 205, Puebla 72760, Mexico.
| | - María E Meneses
- Centro de Biotecnología de Hongos Comestibles, Funcionales y Medicinales (CB-HCFM), Campus Puebla, Colegio de Postgraduados (CP), Boulevard Forjadores de Puebla no. 205, Puebla 72760, Mexico.
- CONACYT-Colegio de Postgraduados (CP), Campus Puebla, Boulevard Forjadores de Puebla 205, Puebla 72760, Mexico
| | - Mónica Sánchez-Tapia
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Mexico City 14080, Mexico
| | - Daniel Pérez-Luna
- Centro de Biotecnología de Hongos Comestibles, Funcionales y Medicinales (CB-HCFM), Campus Puebla, Colegio de Postgraduados (CP), Boulevard Forjadores de Puebla no. 205, Puebla 72760, Mexico.
| | - Nimbe Torres
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Mexico City 14080, Mexico
| | - Iván Torre-Villalvazo
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Mexico City 14080, Mexico
| | - Myrna Bonilla
- Centro de Biotecnología de Hongos Comestibles, Funcionales y Medicinales (CB-HCFM), Campus Puebla, Colegio de Postgraduados (CP), Boulevard Forjadores de Puebla no. 205, Puebla 72760, Mexico.
| | - Beatriz Petlacalco
- Centro de Biotecnología de Hongos Comestibles, Funcionales y Medicinales (CB-HCFM), Campus Puebla, Colegio de Postgraduados (CP), Boulevard Forjadores de Puebla no. 205, Puebla 72760, Mexico.
| | - Ivan Castillo
- Centro de Biotecnología de Hongos Comestibles, Funcionales y Medicinales (CB-HCFM), Campus Puebla, Colegio de Postgraduados (CP), Boulevard Forjadores de Puebla no. 205, Puebla 72760, Mexico.
| | - Adriana López-Barradas
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Mexico City 14080, Mexico
| | - Antonio Macías
- Centro de Biotecnología de Hongos Comestibles, Funcionales y Medicinales (CB-HCFM), Campus Puebla, Colegio de Postgraduados (CP), Boulevard Forjadores de Puebla no. 205, Puebla 72760, Mexico.
| | - Armando R Tovar
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Mexico City 14080, Mexico
| | - Daniel Martínez-Carrera
- Centro de Biotecnología de Hongos Comestibles, Funcionales y Medicinales (CB-HCFM), Campus Puebla, Colegio de Postgraduados (CP), Boulevard Forjadores de Puebla no. 205, Puebla 72760, Mexico.
| |
Collapse
|
6
|
Yang D, Kim JW, Jeong H, Kim MS, Lim CW, Lee K, Kim B. Effects of maternal cigarette smoke exposure on the progression of nonalcoholic steatohepatitis in offspring mice. Toxicol Res 2023; 39:91-103. [PMID: 36726830 PMCID: PMC9839905 DOI: 10.1007/s43188-022-00153-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/01/2022] [Accepted: 08/22/2022] [Indexed: 02/04/2023] Open
Abstract
Cigarette smoke (CS) is a dominant carcinogenic agent in a variety of human cancers. CS exposure during pregnancy can adversely affect the fetus. Non-alcoholic fatty liver disease (NAFLD) is considered as a hepatic manifestation of a metabolic disorder, and ranges from simple steatosis to cirrhosis leading to hepatocellular carcinoma. Non-alcoholic steatohepatitis (NASH) is a more severe phase of NAFLD. Recently, there is increasing apprehension about the CS-related chronic liver diseases. Therefore, we examined whether maternal CS exposure could affect the pathogenesis of NASH in offspring. Mainstream CS (MSCS) was exposed to pregnant C57BL/6 mice via nose-only inhalation for 2 h/day, 5 days/week for 2 weeks from day 6 to 17 of gestation at 0, 300, or 600 μg/L. Three-week-old male offspring mice were fed methionine and choline-supplemented (MCS) diet or methionine and choline-deficient including high-fat (MCDHF) diet for 6 weeks to induce NASH. Maternal MSCS exposure increased the severity of NASH by increasing serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels, hepatic total cholesterol (TC) and triglyceride (TG) levels, pro-inflammation, fibrosis, and steatosis in offspring mice. Especially, maternal MSCS exposure significantly downregulated the phosphorylation of AMP-activated protein kinase (AMPK) in MCDHF diet-fed offspring mice. Subsequently, the protein levels of sterol regulatory element-binding protein (SREBP)-1c and stearoyl-CoA desaturase-1 (SCD1) were upregulated by maternal MSCS exposure. In conclusion, maternal MSCS exposure exacerbates the progression of NASH by modulating lipogenesis on offspring mice. Supplementary Information The online version contains supplementary material available at 10.1007/s43188-022-00153-1.
Collapse
Affiliation(s)
- Daram Yang
- Biosafety Research Institute and Laboratory of Veterinary Pathology, College of Veterinary Medicine, Jeonbuk National University, 79 Gobong-Ro, Iksan-Si, Jeollabuk-Do 54596 Republic of Korea
| | - Jong Won Kim
- Biosafety Research Institute and Laboratory of Veterinary Pathology, College of Veterinary Medicine, Jeonbuk National University, 79 Gobong-Ro, Iksan-Si, Jeollabuk-Do 54596 Republic of Korea
| | - Hyuneui Jeong
- Biosafety Research Institute and Laboratory of Veterinary Pathology, College of Veterinary Medicine, Jeonbuk National University, 79 Gobong-Ro, Iksan-Si, Jeollabuk-Do 54596 Republic of Korea
| | - Min Seok Kim
- Inhalation Toxicology Center, Jeonbuk Department of Inhalation Research, Korea Institute of Toxicology, 30, Baekak 1-Gil, Jeongeup, 56212 Republic of Korea
| | - Chae Woong Lim
- Biosafety Research Institute and Laboratory of Veterinary Pathology, College of Veterinary Medicine, Jeonbuk National University, 79 Gobong-Ro, Iksan-Si, Jeollabuk-Do 54596 Republic of Korea
| | - Kyuhong Lee
- Inhalation Toxicology Center, Jeonbuk Department of Inhalation Research, Korea Institute of Toxicology, 30, Baekak 1-Gil, Jeongeup, 56212 Republic of Korea
| | - Bumseok Kim
- Biosafety Research Institute and Laboratory of Veterinary Pathology, College of Veterinary Medicine, Jeonbuk National University, 79 Gobong-Ro, Iksan-Si, Jeollabuk-Do 54596 Republic of Korea
| |
Collapse
|
7
|
Yen CC, Lii CK, Chen CC, Li CC, Tseng MH, Lo CW, Liu KL, Yang YC, Chen HW. Andrographolide Inhibits Lipotoxicity-Induced Activation of the NLRP3 Inflammasome in Bone Marrow-Derived Macrophages. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2022; 51:129-147. [PMID: 36419253 DOI: 10.1142/s0192415x23500088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Andrographolide is the major bioactive component of the herb Andrographis paniculata and is a potent anti-inflammatory agent. Obesity leads to an excess of free fatty acids, particularly palmitic acid (PA), in the circulation. Obesity also causes the deposition of ectopic fat in nonadipose tissues, which leads to lipotoxicity, a condition closely associated with inflammation. Here, we investigated whether andrographolide could inhibit PA-induced inflammation by activating autophagy, activating the antioxidant defense system, and blocking the activation of the NLRP3 inflammasome. Bone marrow-derived macrophages (BMDMs) were primed with lipopolysaccharide (LPS) and then activated with PA. LPS/PA treatment increased both the mRNA expression of NLRP3 and IL-1[Formula: see text] and the release of IL-1[Formula: see text] in BMDMs. Andrographolide inhibited the LPS/PA-induced protein expression of caspase-1 and the release of IL-1[Formula: see text]. Furthermore, andrographolide attenuated LPS/PA-induced mtROS generation by first promoting autophagic flux and catalase activity, and ultimately inhibiting activation of the NLRP3 inflammasome. Our results suggest that the mechanisms by which andrographolide downregulates LPS/PA-induced IL-1[Formula: see text] release in BMDMs involve promoting autophagic flux and catalase activity. Andrographolide may thus be a candidate to prevent obesity- and lipotoxicity-driven chronic inflammatory disease.
Collapse
Affiliation(s)
- Chih-Ching Yen
- Department of Respiratory Therapy, China Medical University, Taichung, Taiwan.,Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Chong-Kuei Lii
- Department of Nutrition, China Medical University, Taichung, Taiwan.,Department of Food Nutrition and Health Biotechnology, Asia University, Taichung, Taiwan
| | - Chih-Chieh Chen
- Department of Nutrition, China Medical University, Taichung, Taiwan
| | - Chien-Chun Li
- Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan.,Department of Nutrition, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Meng-Hsien Tseng
- Department of Nutrition, China Medical University, Taichung, Taiwan
| | - Chia-Wen Lo
- Department of Nutrition, China Medical University, Taichung, Taiwan
| | - Kai-Li Liu
- Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan.,Department of Nutrition, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Ya-Chen Yang
- Department of Food Nutrition and Health Biotechnology, Asia University, Taichung, Taiwan
| | - Haw-Wen Chen
- Department of Nutrition, China Medical University, Taichung, Taiwan
| |
Collapse
|
8
|
Golinska MA, Stubbs M, Harris AL, Boros LG, Basetti M, McIntyre DJO, Griffiths JR. Survival Pathways of HIF-Deficient Tumour Cells: TCA Inhibition, Peroxisomal Fatty Acid Oxidation Activation and an AMPK-PGC-1α Hypoxia Sensor. Cells 2022; 11:3595. [PMID: 36429023 PMCID: PMC9688062 DOI: 10.3390/cells11223595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/09/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
The HIF-1 and HIF-2 (HIF1/2) hypoxia responses are frequently upregulated in cancers, and HIF1/2 inhibitors are being developed as anticancer drugs. How could cancers resist anti-HIF1/2 therapy? We studied metabolic and molecular adaptations of HIF-1β-deficient Hepa-1c4, a hepatoma model lacking HIF1/2 signalling, which mimics a cancer treated by a totally effective anti-HIF1/2 agent. [1,2-13C2]-D-glucose metabolism was measured by SiDMAP metabolic profiling, gene expression by TaqMan, and metabolite concentrations by 1H MRS. HIF-1β-deficient Hepa-1c4 responded to hypoxia by increasing glucose uptake and lactate production. They showed higher glutamate, pyruvate dehydrogenase, citrate shuttle, and malonyl-CoA fluxes than normal Hepa-1 cells, whereas pyruvate carboxylase, TCA, and anaplerotic fluxes decreased. Hypoxic HIF-1β-deficient Hepa-1c4 cells increased expression of PGC-1α, phospho-p38 MAPK, and PPARα, suggesting AMPK pathway activation to survive hypoxia. They had higher intracellular acetate, and secreted more H2O2, suggesting increased peroxisomal fatty acid β-oxidation. Simultaneously increased fatty acid synthesis and degradation would have "wasted" ATP in Hepa-1c4 cells, thus raising the [AMP]:[ATP] ratio, and further contributing to the upregulation of the AMPK pathway. Since these tumour cells can proliferate without the HIF-1/2 pathways, combinations of HIF1/2 inhibitors with PGC-1α or AMPK inhibitors should be explored.
Collapse
Affiliation(s)
- Monika A. Golinska
- Cancer Research UK Cambridge Institute, Cambridge University, Li Ka Shing Centre, Cambridge CB2 0RE, UK
| | - Marion Stubbs
- Cancer Research UK Cambridge Institute, Cambridge University, Li Ka Shing Centre, Cambridge CB2 0RE, UK
| | - Adrian L. Harris
- Hypoxia and Angiogenesis Group, Weatherall Institute of Molecular Medicine, Department of Oncology, University of Oxford, Oxford OX3 9DS, UK
| | - Laszlo G. Boros
- Department of Pediatrics, Harbor-UCLA Medical Center, University of California Los Angeles School of Medicine, Los Angeles, CA 90502, USA
- SiDMAP, LLC, and the Deutenomics Science Institute, 2990 S. Sepulveda BLVD. #300B, Culver City, CA 90064, USA
- The Lundquist Institute for Biomedical Innovation at the Harbor-UCLA Medical Center, 1124 W Carson St, Torrance, CA 90502, USA
- Submolecular Medical Sciences, Vrije University of Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Madhu Basetti
- Cancer Research UK Cambridge Institute, Cambridge University, Li Ka Shing Centre, Cambridge CB2 0RE, UK
| | - Dominick J. O. McIntyre
- Cancer Research UK Cambridge Institute, Cambridge University, Li Ka Shing Centre, Cambridge CB2 0RE, UK
| | - John R. Griffiths
- Cancer Research UK Cambridge Institute, Cambridge University, Li Ka Shing Centre, Cambridge CB2 0RE, UK
| |
Collapse
|
9
|
Lu Z, Sun GF, Pan XA, Qu XH, Yang P, Chen ZP, Han XJ, Wang T. BCATc inhibitor 2 ameliorated mitochondrial dysfunction and apoptosis in oleic acid-induced non-alcoholic fatty liver disease model. Front Pharmacol 2022; 13:1025551. [PMID: 36386234 PMCID: PMC9650408 DOI: 10.3389/fphar.2022.1025551] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/18/2022] [Indexed: 09/14/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a prevalent hepatic disease in the world. Disorders of branched chain amino acid (BCAA) metabolism is involved in various diseases. In this study, we aim to explore the role of BCAA metabolism in the development of NAFLD and the protective effect of BCATc Inhibitor 2, an inhibitor of cytosolic branched chain amino acid transaminase, against NAFLD as well as its underlying mechanism. It was found that oleic acid induced lipid accumulation and apoptosis in HepG2 and LO2 cells. Supplementation of BCAAs further aggravated oleic acid-induced lipid accumulation and apoptosis. In contrast, treatment of BCATc Inhibitor 2 ameliorated oleic acid-induced lipid accumulation and apoptosis. Molecularly, supplementation of BCAAs or treatment of BCATc Inhibitor 2 up-regulated or down-regulated the expression of SREBP1 and lipogenesis-related genes without affecting lipolysis-related genes. BCATc Inhibitor 2 maintained mitochondrial function by ameliorating oleic acid-induced mitochondrial ROS generation and mitochondrial membrane potential disruption. In addition, BCATc Inhibitor 2 treatment alleviated oleic acid-induced activation of JNK and AKT signaling pathway and Bcl2/Bax/Caspase axis. In conclusion, our results indicate BCAA metabolism is involved in NAFLD and BCATc Inhibitor 2 protects against oleic acid-induced lipid accumulation and apoptosis. These findings suggest that BCATc Inhibitor 2 is a promising candidate drug for the treatment of NAFLD.
Collapse
Affiliation(s)
- Zhuo Lu
- Institute of Geriatrics, Jiangxi Provincial People’s Hospital and The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Gui-Feng Sun
- Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, China
| | - Xiao-An Pan
- Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, China
| | - Xin-Hui Qu
- Institute of Geriatrics, Jiangxi Provincial People’s Hospital and The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- Department of Neurology, Jiangxi Provincial People’s Hospital and The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Ping Yang
- Department of Neurology, Jiangxi Provincial People’s Hospital and The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Zhi-Ping Chen
- Department of Critical Care Medicine, Jiangxi Provincial People’s Hospital and The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Xiao-Jian Han
- Institute of Geriatrics, Jiangxi Provincial People’s Hospital and The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, China
- Department of Neurology, Jiangxi Provincial People’s Hospital and The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Tao Wang
- Institute of Geriatrics, Jiangxi Provincial People’s Hospital and The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| |
Collapse
|
10
|
Castellanos DB, Martín-Jiménez CA, Pinzón A, Barreto GE, Padilla-González GF, Aristizábal A, Zuluaga M, González Santos J. Metabolomic Analysis of Human Astrocytes in Lipotoxic Condition: Potential Biomarker Identification by Machine Learning Modeling. Biomolecules 2022; 12:biom12070986. [PMID: 35883542 PMCID: PMC9313230 DOI: 10.3390/biom12070986] [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: 05/03/2022] [Revised: 06/15/2022] [Accepted: 06/27/2022] [Indexed: 02/04/2023] Open
Abstract
The association between neurodegenerative diseases (NDs) and obesity has been well studied in recent years. Obesity is a syndrome of multifactorial etiology characterized by an excessive accumulation and release of fatty acids (FA) in adipose and non-adipose tissue. An excess of FA generates a metabolic condition known as lipotoxicity, which triggers pathological cellular and molecular responses, causing dysregulation of homeostasis and a decrease in cell viability. This condition is a hallmark of NDs, and astrocytes are particularly sensitive to it, given their crucial role in energy production and oxidative stress management in the brain. However, analyzing cellular mechanisms associated with these conditions represents a challenge. In this regard, metabolomics is an approach that allows biochemical analysis from the comprehensive perspective of cell physiology. This technique allows cellular metabolic profiles to be determined in different biological contexts, such as those of NDs and specific metabolic insults, including lipotoxicity. Since data provided by metabolomics can be complex and difficult to interpret, alternative data analysis techniques such as machine learning (ML) have grown exponentially in areas related to omics data. Here, we developed an ML model yielding a 93% area under the receiving operating characteristic (ROC) curve, with sensibility and specificity values of 80% and 93%, respectively. This study aimed to analyze the metabolomic profiles of human astrocytes under lipotoxic conditions to provide powerful insights, such as potential biomarkers for scenarios of lipotoxicity induced by palmitic acid (PA). In this work, we propose that dysregulation in seleno-amino acid metabolism, urea cycle, and glutamate metabolism pathways are major triggers in astrocyte lipotoxic scenarios, while increased metabolites such as alanine, adenosine, and glutamate are suggested as potential biomarkers, which, to our knowledge, have not been identified in human astrocytes and are proposed as candidates for further research and validation.
Collapse
Affiliation(s)
- Daniel Báez Castellanos
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110311, Colombia; (D.B.C.); (A.A.)
| | - Cynthia A. Martín-Jiménez
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA 30329-4208, USA;
| | - Andrés Pinzón
- Laboratorio de Bioinformática y Biología de Sistemas, Universidad Nacional de Colombia, Bogotá 111321, Colombia;
| | - George E. Barreto
- Department of Biological Sciences, University of Limerick, V94 T9PX Limerick, Ireland;
| | | | - Andrés Aristizábal
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110311, Colombia; (D.B.C.); (A.A.)
| | - Martha Zuluaga
- Escuela de Ciencias Básicas Tecnologías e Ingenierías, Universidad Nacional Abierta y a Distancia, Dosquebradas 661001, Colombia;
| | - Janneth González Santos
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110311, Colombia; (D.B.C.); (A.A.)
- Correspondence: ; Tel.: +57-60-1-3208320
| |
Collapse
|
11
|
Valencia FP, Marino AF, Noutsos C, Poon K. Concentration-dependent change in hypothalamic neuronal transcriptome by the dietary fatty acids: oleic and palmitic acids. J Nutr Biochem 2022; 106:109033. [DOI: 10.1016/j.jnutbio.2022.109033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 12/20/2021] [Accepted: 03/18/2022] [Indexed: 11/30/2022]
|
12
|
Kim MJ, Kim JH, Lee S, Kim B, Kim HY. The protective effects of Aster yomena (Kitam.) Honda on high-fat diet-induced obese C57BL/6J mice. Nutr Res Pract 2022; 16:46-59. [PMID: 35116127 PMCID: PMC8784267 DOI: 10.4162/nrp.2022.16.1.46] [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: 08/05/2021] [Revised: 10/07/2021] [Accepted: 11/23/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND/OBJECTIVES Aster yomena (Kitam.) Honda (AY) has remarkable bioactivities, such as antioxidant, anti-inflammation, and anti-cancer activities. On the other hand, the effects of AY against obesity-induced insulin resistance have not been reported. Therefore, this study examined the potential of AY against obesity-associated insulin resistance in high-fat diet (HFD)-fed mice. MATERIALS/METHODS An obesity model was established by feeding C57BL/6J mice a 60% HFD for 16 weeks. The C57BL6/When ethyl acetate fraction from AY (EFAY) at doses of 100 and 200 mg/kg/day was administered orally to mice fed a HFD for the last 4 weeks. Normal and control groups were administered water orally. The body weight and fasting blood glucose were measured every week. Dietary intake was measured every other day. After dissection, blood and tissues were collected from the mice. RESULTS The administration of EFAY reduced body and organ weights significantly compared to HFD-fed control mice. The EFAY-administered groups also improved the serum lipid profile by decreasing the triglyceride, total cholesterol, and low-density lipoprotein compared to the control group. In addition, EFAY ameliorated the insulin resistance-related metabolic dysfunctions, including the fasting blood glucose and serum insulin level, compared to the HFD-fed control mice. The EFAY inhibited lipid synthesis and insulin resistance by down-regulation of hepatic fatty acid synthase and up-regulation of the AMP-activated protein kinase pathway. EFAY also reduced lipid peroxidation in the liver, indicating that EFAY protected hepatic injury induced by obesity. CONCLUSIONS These results suggest that EFAY improved obesity-associated insulin resistance by regulating the lipid and glucose metabolism, suggesting that AY could be used as a functional food to prevent obesity and insulin resistance.
Collapse
Affiliation(s)
- Min Jeong Kim
- Department of Food Science and Nutrition, Pusan National University, Busan 46241, Korea
| | - Ji Hyun Kim
- Department of Food Science, Gyeongsang National University, Jinju 52725, Korea
| | - Sanghyun Lee
- Department of Plant Science and Technology, Chung-Ang University, Anseong 17546, Korea
| | - Bohkyung Kim
- Department of Food Science and Nutrition, Pusan National University, Busan 46241, Korea
| | - Hyun Young Kim
- Department of Food Science, Gyeongsang National University, Jinju 52725, Korea
| |
Collapse
|
13
|
Xi Y, Zheng J, Xie W, Xu X, Cho N, Zhou X, Yu X. (+)-Dehydrovomifoliol Alleviates Oleic Acid-Induced Lipid Accumulation in HepG2 Cells via the PPARα-FGF21 Pathway. Front Pharmacol 2021; 12:750147. [PMID: 34867358 PMCID: PMC8640464 DOI: 10.3389/fphar.2021.750147] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/20/2021] [Indexed: 11/13/2022] Open
Abstract
An overload of hepatic fatty acids, such as oleic acid is a key trigger of non-alcoholic fatty liver disease (NAFLD). Here, we investigated whether Artemisia frigida, a valuable traditional medicine used to treat various diseases, could mitigate OA-induced lipid accumulation in HepG2 cells. Then, to identify the active substances in A. frigida, a phytochemistry investigation was conducted using a bioassay-guided isolation method. Consequently, one terpene (1) and one flavone (2) were identified. Compound 1 ((+)-dehydrovomifoliol) exhibited potent effects against lipid accumulation in OA-induced HepG2 cells, without causing cyto-toxicity. Notably, treatment with (+)-dehydrovomifoliol decreased the expression levels of three genes related to lipogenesis (SREBP1, ACC, and FASN) and increased those of three genes related to fatty acid oxidation (PPARα, ACOX1, and FGF21). In addition, similar results were observed for SREBP1, PPARα, and FGF21 protein levels. The effects of (+)-dehydrovomifoliol were partially reversed by treatment with the PPARα antagonist GW6471, indicating the important role of the PPARα-FGF21 axis in the effects of (+)-dehydrovomifoliol. Based on its effects on hepatic lipogenesis and fatty acid oxidation signaling via the PPARα-FGF21 axis, (+)-dehydrovomifoliol isolated from A. frigida could be a useful early lead compound for developing new drugs for NAFLD prevention.
Collapse
Affiliation(s)
- Yiyuan Xi
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju, Korea
| | - Jujia Zheng
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wei Xie
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiangwei Xu
- Pharmacy Department, Yongkang First People's Hospital, Jinhua, China
| | - Namki Cho
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju, Korea
| | - Xudong Zhou
- TCM and Ethnomedicine Innovation and Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Xiaomin Yu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
14
|
Costa A, Reynés B, Konieczna J, Martín M, Fiol M, Palou A, Romaguera D, Oliver P. Use of human PBMC to analyse the impact of obesity on lipid metabolism and metabolic status: a proof-of-concept pilot study. Sci Rep 2021; 11:18329. [PMID: 34526523 PMCID: PMC8443582 DOI: 10.1038/s41598-021-96981-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 08/18/2021] [Indexed: 12/12/2022] Open
Abstract
Peripheral blood mononuclear cells (PBMC) are widely used as a biomarker source in nutrition/obesity studies because they reflect gene expression profiles of internal tissues. In this pilot proof-of-concept study we analysed in humans if, as we previously suggested in rodents, PBMC could be a surrogate tissue to study overweight/obesity impact on lipid metabolism. Pre-selected key lipid metabolism genes based in our previous preclinical studies were analysed in PBMC of normoglycemic normal-weight (NW), and overweight-obese (OW-OB) subjects before and after a 6-month weight-loss plan. PBMC mRNA levels of CPT1A, FASN and SREBP-1c increased in the OW-OB group, according with what described in liver and adipose tissue of humans with obesity. This altered expression pattern was related to increased adiposity and early signs of metabolic impairment. Greater weight loss and/or metabolic improvement as result of the intervention was related to lower CPT1A, FASN and SREBP-1c gene expression in an adjusted linear mixed-effects regression analysis, although no gene expression recovery was observed when considering mean comparisons. Thus, human PBMC reflect lipid metabolism expression profile of energy homeostatic tissues, and early obesity-related alterations in metabolic at-risk subjects. Further studies are needed to understand PBMC usefulness for analysis of metabolic recovery in weigh management programs.
Collapse
Affiliation(s)
- Andrea Costa
- Nutrigenomics, Biomarkers and Risk Evaluation (NuBE) Group, University of the Balearic Islands (UIB), Palma, Spain.,Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.,CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Madrid, Spain
| | - Bàrbara Reynés
- Nutrigenomics, Biomarkers and Risk Evaluation (NuBE) Group, University of the Balearic Islands (UIB), Palma, Spain.,Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.,CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Madrid, Spain
| | - Jadwiga Konieczna
- Research Group on Nutritional Epidemiology and Cardiovascular Physiopathology (NUTRECOR), University Hospital Son Espases (HUSE), Palma, Spain.,Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.,CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Madrid, Spain
| | - Marian Martín
- Research Group on Nutritional Epidemiology and Cardiovascular Physiopathology (NUTRECOR), University Hospital Son Espases (HUSE), Palma, Spain.,Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.,CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Madrid, Spain
| | - Miquel Fiol
- Research Group on Nutritional Epidemiology and Cardiovascular Physiopathology (NUTRECOR), University Hospital Son Espases (HUSE), Palma, Spain.,Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.,CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Madrid, Spain
| | - Andreu Palou
- Nutrigenomics, Biomarkers and Risk Evaluation (NuBE) Group, University of the Balearic Islands (UIB), Palma, Spain. .,Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain. .,CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Madrid, Spain.
| | - Dora Romaguera
- Research Group on Nutritional Epidemiology and Cardiovascular Physiopathology (NUTRECOR), University Hospital Son Espases (HUSE), Palma, Spain.,Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.,CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Madrid, Spain
| | - Paula Oliver
- Nutrigenomics, Biomarkers and Risk Evaluation (NuBE) Group, University of the Balearic Islands (UIB), Palma, Spain.,Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.,CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Madrid, Spain
| |
Collapse
|
15
|
Ghnaimawi S, Rebello L, Baum J, Huang Y. DHA but not EPA induces the trans-differentiation of C2C12 cells into white-like adipocytes phenotype. PLoS One 2021; 16:e0249438. [PMID: 34473703 PMCID: PMC8412409 DOI: 10.1371/journal.pone.0249438] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 08/12/2021] [Indexed: 01/21/2023] Open
Abstract
Muscle derived stem cells (MDSCs) and myoblast play an important role in myotube regeneration when muscle tissue is injured. However, these cells can be induced to differentiate into adipocytes once exposed to PPARγ activator like EPA and DHA that are highly suggested during pregnancy. The objective of this study aims at determining the identity of trans-differentiated cells by exploring the effect of EPA and DHA on C2C12 undergoing differentiation into brown and white adipocytes. DHA but not EPA committed C2C12 cells reprograming into white like adipocyte phenotype. Also, DHA promoted the expression of lipolysis regulating genes but had no effect on genes regulating β-oxidation referring to its implication in lipid re-esterification. Furthermore, DHA impaired C2C12 cells differentiation into brown adipocytes through reducing the thermogenic capacity and mitochondrial biogenesis of derived cells independent of UCP1. Accordingly, DHA treated groups showed an increased accumulation of lipid droplets and suppressed mitochondrial maximal respiration and spare respiratory capacity. EPA, on the other hand, reduced myogenesis regulating genes, but no significant differences were observed in the expression of adipogenesis key genes. Likewise, EPA suppressed the expression of WAT signature genes indicating that EPA and DHA have an independent role on white adipogensis. Unlike DHA treatment, EPA supplementation had no effect on the differential of C2C12 cells into brown adipocytes. In conclusion, DHA is a potent adipogenic and lipogenic factor that can change the metabolic profile of muscle cells by increasing myocellular fat.
Collapse
Affiliation(s)
- Saeed Ghnaimawi
- Department of Cell and Molecular Biology Program, University of Arkansas, Fayetteville, North Carolina, United States of America
| | - Lisa Rebello
- Department of Cell and Molecular Biology Program, University of Arkansas, Fayetteville, North Carolina, United States of America
| | - Jamie Baum
- Department of Food Science, Division of Agriculture, University of Arkansas, Fayetteville, AR, United States of America
| | - Yan Huang
- Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, North Carolina, United States of America
| |
Collapse
|
16
|
Insights into the Cellular and Molecular Mechanisms That Govern the Fracture-Healing Process: A Narrative Review. J Clin Med 2021; 10:jcm10163554. [PMID: 34441849 PMCID: PMC8397080 DOI: 10.3390/jcm10163554] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 08/07/2021] [Accepted: 08/10/2021] [Indexed: 12/28/2022] Open
Abstract
Fracture-healing is a complex multi-stage process that usually progresses flawlessly, resulting in restoration of bone architecture and function. Regrettably, however, a considerable number of fractures fail to heal, resulting in delayed unions or non-unions. This may significantly impact several aspects of a patient’s life. Not surprisingly, in the past few years, a substantial amount of research and number of clinical studies have been designed, aiming at shedding light into the cellular and molecular mechanisms that regulate fracture-healing. Herein, we present the current knowledge on the pathobiology of the fracture-healing process. In addition, the role of skeletal cells and the impact of marrow adipose tissue on bone repair is discussed. Unveiling the pathogenetic mechanisms that govern the fracture-healing process may lead to the development of novel, smarter, and more effective therapeutic strategies for the treatment of fractures, especially of those with large bone defects.
Collapse
|
17
|
Lhomme T, Clasadonte J, Imbernon M, Fernandois D, Sauve F, Caron E, Lima N, Heras V, Martinez-Corral I, Müller-Fielitz H, Rasika S, Schwaninger M, Nogueiras R, Prevot V. Tanycytic networks mediate energy balance by feeding lactate to glucose-insensitive POMC neurons. J Clin Invest 2021; 131:e140521. [PMID: 34324439 DOI: 10.1172/jci140521] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/28/2021] [Indexed: 11/17/2022] Open
Abstract
Hypothalamic glucose sensing enables an organism to match energy expenditure and food intake to circulating levels of glucose, the main energy source of the brain. Here, we established that tanycytes of the hypothalamic arcuate nucleus, specialized glia that line the wall of the third ventricle, convert brain glucose supplies into lactate that they transmit through monocarboxylate transporters to arcuate proopiomelanocortin neurons, which integrate this signal to drive their activity and to adapt the metabolic response to meet physiological demands. Furthermore, this transmission required the formation of extensive Connexin-43 gap-junction-mediated metabolic networks by arcuate tanycytes. Selectively suppressing either tanycytic monocarboxylate transporters or gap junctions resulted in altered feeding behavior and energy metabolism. Tanycytic intercellular communication and lactate production are thus integral to the mechanism by which hypothalamic neurons that regulate energy and glucose homeostasis efficiently perceive alterations in systemic glucose levels as a function of the physiological state of the organism.
Collapse
Affiliation(s)
- Tori Lhomme
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Jerome Clasadonte
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Monica Imbernon
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Daniela Fernandois
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Florent Sauve
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Emilie Caron
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Natalia Lima
- CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Violeta Heras
- CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ines Martinez-Corral
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Helge Müller-Fielitz
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - S Rasika
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Ruben Nogueiras
- CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Vincent Prevot
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| |
Collapse
|
18
|
Yang ZY, Chen WL. Examining the Association Between Serum Leptin and Sarcopenic Obesity. J Inflamm Res 2021; 14:3481-3487. [PMID: 34326656 PMCID: PMC8315286 DOI: 10.2147/jir.s320445] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 07/12/2021] [Indexed: 01/02/2023] Open
Abstract
Objective Sarcopenic obesity is an emerging geriatric syndrome among elderly individuals. Studies revealed a complicated pathogenesis between sarcopenia and obesity. Leptin, a proinflammatory adipokine, has been implicated in the mechanism of sarcopenic obesity. This study investigated the relationship between serum leptin level and sarcopenic obesity. Methods The study included 4063 participants aged 60 years and older from the NHANES III database. Sarcopenia was defined as a skeletal muscle index (SMI) less than one standard deviation below the young adult value. Obesity was defined as a body mass index (BMIF) over 30 kg/m2. Multivariate regression analysis was performed to examine the association between serum leptin level and sarcopenic obesity. Results In adjusted models, serum leptin level was positively correlated with BMI (β: 1.33, p value < 0.001) and negatively correlated with SMI (β: −0.091, p value = 0.001). A significant association between serum leptin level and sarcopenic obesity was found in multivariate analysis (β: 4.011, p value=0.014). Conclusion Our study demonstrated that serum leptin level was related to an increased risk of sarcopenic obesity. This epidemiologic finding suggests that leptin may play a role in sarcopenic obesity.
Collapse
Affiliation(s)
- Zhe-Yu Yang
- Department of General Medicine, Tri-Service General Hospital; and School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Division of Family Medicine, Department of Family and Community Medicine, Tri-Service General Hospital; and School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Wei-Liang Chen
- Division of Family Medicine, Department of Family and Community Medicine, Tri-Service General Hospital; and School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital; and School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Department of Biochemistry, National Defense Medical Center, Taiwan, Republic of China
| |
Collapse
|
19
|
Mahat B, Mauger JF, Imbeault P. Effects of different oxygen tensions on differentiated human preadipocytes lipid storage and mobilisation. Arch Physiol Biochem 2021; 127:37-43. [PMID: 31055955 DOI: 10.1080/13813455.2019.1609995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Adipose tissue expansion has been suggested to impair oxygen (O2) diffusion in the adipose tissue and cause hypoxia. This study aimed at characterising the effects of hypoxia on adipocyte lipid storage and mobilisation functions. Human preadipocytes were exposed to different O2 tensions (3, 10 and 21%) either acutely for 24 h after differentiation (acute exposure) or during differentiation (14d, chronic hypoxia). Lipoprotein lipase (LPL) activity was decreased dose-dependently by both acute and chronic hypoxia (p < .05). Acute exposure to 3, and 10% O2 stimulated the expression of lipid storage gene, while chronic exposure to 3% O2 inhibited the expression of genes involved in lipid storage and mobilisation (p < .05). Acute hypoxia dose-dependently stimulated basal lipolysis. Conversely, chronic hypoxia did not affect basal lipolysis but significantly decreased isoproterenol-stimulated lipolysis (p < .05). In conclusion, the effects of hypoxia on human adipocyte lipid storage and mobilisation functions are complex but could favour ectopic fat deposition.
Collapse
Affiliation(s)
- Bimit Mahat
- Behavioral and Metabolic Research Unit, School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, Canada
| | - Jean-François Mauger
- Behavioral and Metabolic Research Unit, School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, Canada
| | - Pascal Imbeault
- Behavioral and Metabolic Research Unit, School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, Canada
- Institut du savoir Montfort, Hôpital Montfort, Ottawa, Canada
| |
Collapse
|
20
|
Pan M, Qin C, Han X. Lipid Metabolism and Lipidomics Applications in Cancer Research. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1316:1-24. [PMID: 33740240 PMCID: PMC8287890 DOI: 10.1007/978-981-33-6785-2_1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Lipids are the critical components of cellular and plasma membrane, which constitute an impermeable barrier of cellular compartments, and play important roles on numerous cellular processes including cell growth, proliferation, differentiation, and signaling. Alterations in lipid metabolism have been implicated in the development and progression of cancers. However, unlike other biomolecules, the diversity in the structures and characteristics of lipid species results in the limited understanding of their metabolic alterations in cancers. Lipidomics is an emerging discipline that studies lipids in a large scale based on analytical chemistry principles and technological tools. Multidimensional mass spectrometry-based shotgun lipidomics (MDMS-SL) uses direct infusion to avoid difficulties from alterations in concentration, chromatographic anomalies, and ion-pairing alterations to improve resolution and achieve rapid and accurate qualitative and quantitative analysis. In this chapter, lipids and lipid metabolism relevant to cancer research are introduced, followed by a brief description of MDMS-SL and other shotgun lipidomics techniques and some applications for cancer research.
Collapse
Affiliation(s)
- Meixia Pan
- Barshop Institute for Longevity and Aging Studies, San Antonio, TX, USA
| | - Chao Qin
- Barshop Institute for Longevity and Aging Studies, San Antonio, TX, USA
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, San Antonio, TX, USA.
- Department of Medicine - Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
21
|
Snx14 proximity labeling reveals a role in saturated fatty acid metabolism and ER homeostasis defective in SCAR20 disease. Proc Natl Acad Sci U S A 2020; 117:33282-33294. [PMID: 33310904 DOI: 10.1073/pnas.2011124117] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Fatty acids (FAs) are central cellular metabolites that contribute to lipid synthesis, and can be stored or harvested for metabolic energy. Dysregulation in FA processing and storage causes toxic FA accumulation or altered membrane compositions and contributes to metabolic and neurological disorders. Saturated lipids are particularly detrimental to cells, but how lipid saturation levels are maintained remains poorly understood. Here, we identify the cerebellar ataxia spinocerebellar ataxia, autosomal recessive 20 (SCAR20)-associated protein Snx14, an endoplasmic reticulum (ER)-lipid droplet (LD) tethering protein, as a factor required to maintain the lipid saturation balance of cell membranes. We show that following saturated FA (SFA) treatment, the ER integrity of SNX14 KO cells is compromised, and both SNX14 KO cells and SCAR20 disease patient-derived cells are hypersensitive to SFA-mediated lipotoxic cell death. Using APEX2-based proximity labeling, we reveal the protein composition of Snx14-associated ER-LD contacts and define a functional interaction between Snx14 and Δ-9 FA desaturase SCD1. Lipidomic profiling reveals that SNX14 KO cells increase membrane lipid saturation following exposure to palmitate, phenocopying cells with perturbed SCD1 activity. In line with this, SNX14 KO cells manifest delayed FA processing and lipotoxicity, which can be rescued by SCD1 overexpression. Altogether, these mechanistic insights reveal a role for Snx14 in FA and ER homeostasis, defects in which may underlie the neuropathology of SCAR20.
Collapse
|
22
|
Oyabambi AO, Olaniyi KS, Soladoye AO, Olatunji LA. Suppression of uric acid and lactate production by sodium acetate ameliorates hepatic triglyceride accumulation in fructose-insulin resistant pregnant rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2020; 80:103452. [PMID: 32610186 DOI: 10.1016/j.etap.2020.103452] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 05/30/2020] [Accepted: 06/26/2020] [Indexed: 06/11/2023]
Abstract
High fructose intake has been associated with perturbed lipid, uric acid and lactate homeostasis. However, consumption of fructose-sweetened beverages is not usually regulated during pregnancy. The effect of short-chain fatty acid (acetate) on the metabolic effects of high fructose intake during pregnancy is not known. We hypothesized that acetate prevents gestational fructose-induced hepatic triglyceride (TG) accumulation by suppressing uric acid and lactate production. Pregnant Wistar rats were randomly separated into three groups (n = 6/group) receiving drinking water (CON), 10 % (w/v) fructose drink (FRU) and 10 % (w/v) fructose with 200 mg/kg (w/w; p.o.) sodium acetate (FRU + ACE) daily for nineteen days. Fructose intake resulted in increased body weight gain, liver weight, fluid intake, visceral fat, insulin resistance, fasting blood glucose, insulin, plasma and hepatic TG, total cholesterol, free fatty acid, lipid peroxidation, adenosine deaminase, xanthine oxidase, uric acid, lactate, lactate dehydrogenase, and liver injury marker enzymes. However, gestational high fructose intake led to depressed plasma and hepatic glucose-6-phosphate dehydrogenase (G6PD)-dependent antioxidant barrier, adenosine and food intake. All these effects except water intake and food intake were abated by sodium acetate. These results demonstrate that maternal fructose-enriched drink would cause hepatic TG accumulation that is associated with perturbed glucose, uric acid, lactate homeostasis, and G6PD-dependent antioxidant barrier. These results also demonstrate that acetate protects the liver against gestational fructose-induced TG accumulation by inhibiting uric acid and lactate production. Thus, acetate may be useful in the treatment of hyperuricemia- and hyperlactatemia-related disorders.
Collapse
Affiliation(s)
- Adewumi O Oyabambi
- HOPE Cardiometabolic Research Team & Department of Physiology, Faculty of Basic Medical Sciences, University of Ilorin, Ilorin, Nigeria
| | - Kehinde S Olaniyi
- HOPE Cardiometabolic Research Team & Department of Physiology, Faculty of Basic Medical Sciences, University of Ilorin, Ilorin, Nigeria; Department of Physiology, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Ayodele O Soladoye
- HOPE Cardiometabolic Research Team & Department of Physiology, Faculty of Basic Medical Sciences, University of Ilorin, Ilorin, Nigeria; Department of Physiology, College of Health Sciences, Bowen University, Iwo, Nigeria
| | - Lawrence A Olatunji
- HOPE Cardiometabolic Research Team & Department of Physiology, Faculty of Basic Medical Sciences, University of Ilorin, Ilorin, Nigeria.
| |
Collapse
|
23
|
Sergi D, Williams LM. Potential relationship between dietary long-chain saturated fatty acids and hypothalamic dysfunction in obesity. Nutr Rev 2020; 78:261-277. [PMID: 31532491 DOI: 10.1093/nutrit/nuz056] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Diet-induced hypothalamic inflammation, which leads to hypothalamic dysfunction and a loss of regulation of energy balance, is emerging as a potential driver of obesity. Excessive intake of long-chain saturated fatty acids is held to be the causative dietary component in hypothalamic inflammation. This review summarizes current evidence on the role of long-chain saturated fatty acids in promoting hypothalamic inflammation and the related induction of central insulin and leptin insensitivity. Particularly, the present review focuses on the molecular mechanisms linking long-chain saturated fatty acids and hypothalamic inflammation, emphasizing the metabolic fate of fatty acids and the resulting lipotoxicity, which is a key driver of hypothalamic dysfunction. In conclusion, long-chain saturated fatty acids are key nutrients that promote hypothalamic inflammation and dysfunction by fostering the build-up of lipotoxic lipid species, such as ceramide. Furthermore, when long-chain saturated fatty acids are consumed in combination with high levels of refined carbohydrates, the proinflammatory effects are exacerbated via a mechanism that relies on the formation of advanced glycation end products.
Collapse
Affiliation(s)
- Domenico Sergi
- Nutrition and Health Substantiation Group, Nutrition and Health Program, Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Adelaide, South Australia, Australia
| | - Lynda M Williams
- Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
| |
Collapse
|
24
|
Saiman Y, Hooks R, Carr RM. High-Risk Groups for Non-alcoholic Fatty Liver and Non-alcoholic Steatohepatitis Development and Progression. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/s11901-020-00539-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
25
|
DeBari MK, Abbott RD. Adipose Tissue Fibrosis: Mechanisms, Models, and Importance. Int J Mol Sci 2020; 21:ijms21176030. [PMID: 32825788 PMCID: PMC7503256 DOI: 10.3390/ijms21176030] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/15/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023] Open
Abstract
Increases in adipocyte volume and tissue mass due to obesity can result in inflammation, further dysregulation in adipose tissue function, and eventually adipose tissue fibrosis. Like other fibrotic diseases, adipose tissue fibrosis is the accumulation and increased production of extracellular matrix (ECM) proteins. Adipose tissue fibrosis has been linked to decreased insulin sensitivity, poor bariatric surgery outcomes, and difficulty in weight loss. With the rising rates of obesity, it is important to create accurate models for adipose tissue fibrosis to gain mechanistic insights and develop targeted treatments. This article discusses recent research in modeling adipose tissue fibrosis using in vivo and in vitro (2D and 3D) methods with considerations for biomaterial selections. Additionally, this article outlines the importance of adipose tissue in treating other fibrotic diseases and methods used to detect and characterize adipose tissue fibrosis.
Collapse
Affiliation(s)
- Megan K. DeBari
- Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA;
| | - Rosalyn D. Abbott
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Correspondence:
| |
Collapse
|
26
|
Zhang Z, Yan B, Gao F, Li Q, Meng X, Chen P, Zhou L, Deng W, Li C, Xu W, Han S, Feng H, Li Y, Chen J, Yin Z, Liao C, Tse HF, Xu A, Lian Q. PSCs Reveal PUFA-Provoked Mitochondrial Stress as a Central Node Potentiating RPE Degeneration in Bietti's Crystalline Dystrophy. Mol Ther 2020; 28:2642-2661. [PMID: 32755565 PMCID: PMC7704739 DOI: 10.1016/j.ymthe.2020.07.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/25/2020] [Accepted: 07/21/2020] [Indexed: 12/21/2022] Open
Abstract
Bietti’s crystalline dystrophy (BCD) is an incurable retinal disorder caused by the polypeptide 2 of cytochrome P450 family 4 subfamily V (CYP4V2) mutations. Patients with BCD present degeneration of retinal pigmented epithelial (RPE) cells and consequent blindness. The lack of appropriate disease models and patients’ RPE cells limits our understanding of the pathological mechanism of RPE degeneration. In this study, using CYP4V2 mutant pluripotent stem cells as disease models, we demonstrated that RPE cells with CYP4V2 mutations presented a disrupted fatty acid homeostasis, which were characterized with excessive accumulation of poly-unsaturated fatty acid (PUFA), including arachidonic acid (AA) and eicosapentaenoic acid (EPA). The PUFA overload increased mitochondrial reactive oxygen species, impaired mitochondrial respiratory functions, and triggered mitochondrial stress-activated p53-independent apoptosis in CYP4V2 mutant RPE cells. Restoration of the mutant CYP4V2 using adeno-associated virus 2 (AAV2) can effectively reduce PUFA deposition, alleviate mitochondria oxidative stresses, and rescue RPE cell death in BCD RPE cells. Taken together, our results highlight a role of PUFA-induced mitochondrial damage as a central node to potentiate RPE degeneration in BCD patients. AAV2-mediated gene therapy may represent a feasible strategy for the treatment of BCD.
Collapse
Affiliation(s)
- Zhao Zhang
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; Prenatal Diagnostic Centre and Cord Blood Bank, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Bin Yan
- Department of Computer Science, The University of Hong Kong, Hong Kong SAR, China; School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China; Intervention and Cell Therapy Center, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Fei Gao
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Qing Li
- Ophthalmology, Grantham Hospital, Hospital Authority, Hong Kong SAR, China
| | - Xiaohong Meng
- Southwest Eye Hospital, Third Military Medical University, Chongqing 400038, China
| | - Peikai Chen
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Lei Zhou
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Wen Deng
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Cheng Li
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Weiyi Xu
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Shuo Han
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Hong Feng
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yaping Li
- Ophthalmology, The Second Hospital of Jilin University, Changchun 130022, China
| | - Junhui Chen
- Intervention and Cell Therapy Center, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Zhengqin Yin
- Southwest Eye Hospital, Third Military Medical University, Chongqing 400038, China
| | - Can Liao
- Prenatal Diagnostic Centre and Cord Blood Bank, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Hung-Fat Tse
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| | - Aimin Xu
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China.
| | - Qizhou Lian
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; Prenatal Diagnostic Centre and Cord Blood Bank, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
27
|
Miklishanskaya SV, Solomasova LV, Mazur MA. Obesity and Mechanisms of its Negative Impact on the Cardiovascular System. RATIONAL PHARMACOTHERAPY IN CARDIOLOGY 2020. [DOI: 10.20996/1819-6446-2020-02-09] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Currently, the number of obese people in the world is constantly increasing. Obesity has a direct negative impact on the heart and blood vessels, which can be considered not only as an appropriate response to an increase in the volume of circulating blood due to an increase in body weight, but also as a side tissue reaction of the myocardium to hormonal and metabolic changes inherent in obesity. Our review is devoted to the description of the mechanisms of influence of obesity on the structural and functional parameters of the heart, which create prerequisites for the development of cardiovascular diseases, as well as the existing contradictions. Currently, the accumulated data suggest that an excessive amount of adipose tissue, in addition to metabolic disorders, including insulin resistance, imbalance of adipokines and inflammation markers, leading to the development of lipotoxicity, can directly penetrate the myocardium and cause violations of its contractile properties, as well as affect the conduction of excitation pulses and provoke the development of rhythm and conduction disorders. The development of endothelial dysfunction in obesity ultimately leads to the development of atherosclerosis and coronary heart disease. In addition, obesity contributes to the emergence of risk factors for hypertension, diabetes, atrial fibrillation, chronic heart failure, obstructive sleep apnea syndrome. Given the differences in the literature on the effect of obesity on long-term outcomes in patients with cardiovascular diseases, it is important to conduct prospective studies on the role of individual factors and their combinations that affect the mortality of patients with cardiovascular diseases.
Collapse
Affiliation(s)
| | | | - M. A. Mazur
- Russian Medical Academy of Continuous Professional Education
| |
Collapse
|
28
|
Guevara-Cruz M, Godinez-Salas ET, Sanchez-Tapia M, Torres-Villalobos G, Pichardo-Ontiveros E, Guizar-Heredia R, Arteaga-Sanchez L, Gamba G, Mojica-Espinosa R, Schcolnik-Cabrera A, Granados O, López-Barradas A, Vargas-Castillo A, Torre-Villalvazo I, Noriega LG, Torres N, Tovar AR. Genistein stimulates insulin sensitivity through gut microbiota reshaping and skeletal muscle AMPK activation in obese subjects. BMJ Open Diabetes Res Care 2020; 8:8/1/e000948. [PMID: 32152146 PMCID: PMC7064085 DOI: 10.1136/bmjdrc-2019-000948] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/26/2019] [Accepted: 12/20/2019] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE Obesity is associated with metabolic abnormalities, including insulin resistance and dyslipidemias. Previous studies demonstrated that genistein intake modifies the gut microbiota in mice by selectively increasing Akkermansia muciniphila, leading to reduction of metabolic endotoxemia and insulin sensitivity. However, it is not known whether the consumption of genistein in humans with obesity could modify the gut microbiota reducing the metabolic endotoxemia and insulin sensitivity. RESEARCH DESIGN AND METHODS 45 participants with a Homeostatic Model Assessment (HOMA) index greater than 2.5 and body mass indices of ≥30 and≤40 kg/m2 were studied. Patients were randomly distributed to consume (1) placebo treatment or (2) genistein capsules (50 mg/day) for 2 months. Blood samples were taken to evaluate glucose concentration, lipid profile and serum insulin. Insulin resistance was determined by means of the HOMA for insulin resistance (HOMA-IR) index and by an oral glucose tolerance test. After 2 months, the same variables were assessed including a serum metabolomic analysis, gut microbiota, and a skeletal muscle biopsy was obtained to study the gene expression of fatty acid oxidation. RESULTS In the present study, we show that the consumption of genistein for 2 months reduced insulin resistance in subjects with obesity, accompanied by a modification of the gut microbiota taxonomy, particularly by an increase in the Verrucomicrobia phylum. In addition, subjects showed a reduction in metabolic endotoxemia and an increase in 5'-adenosine monophosphate-activated protein kinase phosphorylation and expression of genes involved in fatty acid oxidation in skeletal muscle. As a result, there was an increase in circulating metabolites of β-oxidation and ω-oxidation, acyl-carnitines and ketone bodies. CONCLUSIONS Change in the gut microbiota was accompanied by an improvement in insulin resistance and an increase in skeletal muscle fatty acid oxidation. Therefore, genistein could be used as a part of dietary strategies to control the abnormalities associated with obesity, particularly insulin resistance; however, long-term studies are needed.
Collapse
Affiliation(s)
- Martha Guevara-Cruz
- Fisiologia de la Nutricion, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico
| | - Einar T Godinez-Salas
- Fisiologia de la Nutricion, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico
| | - Monica Sanchez-Tapia
- Fisiologia de la Nutricion, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico
| | | | - Edgar Pichardo-Ontiveros
- Fisiologia de la Nutricion, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico
| | - Rocio Guizar-Heredia
- Fisiologia de la Nutricion, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico
| | - Liliana Arteaga-Sanchez
- Fisiologia de la Nutricion, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico
| | - Gerardo Gamba
- Nefrología, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico
| | | | | | - Omar Granados
- Fisiologia de la Nutricion, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico
| | - Adriana López-Barradas
- Fisiologia de la Nutricion, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico
| | - Ariana Vargas-Castillo
- Fisiologia de la Nutricion, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico
| | - Ivan Torre-Villalvazo
- Fisiologia de la Nutricion, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico
| | - Lilia G Noriega
- Fisiologia de la Nutricion, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico
| | - Nimbe Torres
- Fisiologia de la Nutricion, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico
| | - Armando R Tovar
- Fisiologia de la Nutricion, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico
| |
Collapse
|
29
|
Sodium acetate protects against nicotine-induced excess hepatic lipid in male rats by suppressing xanthine oxidase activity. Chem Biol Interact 2019; 316:108929. [PMID: 31857090 DOI: 10.1016/j.cbi.2019.108929] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 11/11/2019] [Accepted: 12/15/2019] [Indexed: 12/18/2022]
Abstract
Fatty liver is the hepatic consequence of chronic insulin resistance (IR) and related syndromes. It is mostly accompanied by inflammatory and oxidative molecules. Increased activity of xanthine oxidase (XO) exerts both inflammatory and oxidative effects and has been implicated in metabolic derangements including non-alcoholic fatty liver disease. Short chain fatty acids (SCFAs) elicit beneficial metabolic alterations in IR and related syndromes. In the present study, we evaluated the preventive effects of a SCFA, acetate, on nicotine-induced dysmetabolism and fatty liver. Twenty-four male Wistar rats (n = 6/group): vehicle-treatment (p.o.), nicotine-treated (1.0 mg/kg; p.o.), sodium acetate-treated (200 mg/kg; p.o.) and nicotine + sodium acetate-treated groups. The treatments lasted for 8 weeks. IR was estimated by oral glucose tolerance test and homeostatic model assessment of IR. Plasma and hepatic free fatty acid, triglyceride (TG), glutathione peroxidase, adenosine deaminase (ADA), XO and uric acid (UA) were measured. Nicotine exposure resulted in reduced body weight, liver weight, visceral adiposity, glycogen content and glycogen synthase activity. Conversely, exposure to nicotine increased fasting plasma glucose, lactate, IR, plasma and hepatic TG, free fatty acid, TG/HDL-cholesterol ratio, lipid peroxidation, liver function enzymes, plasma and hepatic UA, XO and ADA activities. However, plasma and hepatic glucose-6-phosphate dehydrogenase-dependent antioxidant defense was not affected by nicotine. Concomitant treatment with acetate ameliorated nicotine-induced effects. Taken together, these results indicate that nicotine exposure leads to excess deposition of lipid in the liver by enhancing XO activity. The results also imply that acetate confers hepatoprotection and is accompanied by decreased XO activity.
Collapse
|
30
|
Shi Y, Zhang W, Cheng Y, Liu C, Chen S. Bromide alleviates fatty acid-induced lipid accumulation in mouse primary hepatocytes through the activation of PPARα signals. J Cell Mol Med 2019; 23:4464-4474. [PMID: 31033195 PMCID: PMC6533524 DOI: 10.1111/jcmm.14347] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 03/15/2019] [Accepted: 04/08/2019] [Indexed: 12/14/2022] Open
Abstract
Increased plasma free fatty acids (FFAs) and liver triglyceride (TG) accumulations have been implicated in the pathogenesis of hepatic steatosis. On the other hand, trace elements function as essential cofactors that are involved in various biochemical processes in mammals, including metabolic homeostasis. Notably, clinical and animal studies suggest that the plasma levels of bromide negatively correlate with those of TG, total cholesterol (TC) and high‐density lipoprotein‐cholesterol (HDL‐C). However, the effect of bromide on lipid accumulation and the direct molecular target responsible for its action remains unknown. Oil red O (ORO) and Nile red staining were used to detect the effect of bromide on lipid accumulation in mouse primary hepatocytes (PHs) treated with different doses of sodium bromide (NaBr) in the presence of FFAs (0.4 mM oleate/palmitic acid 1:1). Spectrophotometric and fluorometric analyses were performed to assess cellular TG concentrations and rates of fatty acid oxidation (FAO), respectively, in mouse PHs. We found that bromide decreased FFA‐induced lipid accumulation and increased FFA‐inhibited oxygen consumptions in mouse PHs in a dose‐dependent manner via activation of PPARα. Mechanical studies demonstrated that bromide decreased the phosphorylation levels of JNK. More importantly, the PPARα‐specific inhibitor GW6471 partially abolished the beneficial effects of bromide on mouse PHs. Bromide alleviates FFA‐induced excessive lipid storage and increases rates of FAO through the activation of PPARα/JNK signals in mouse PHs. Therefore, bromide may serve as a novel drug in the treatment of hepatic steatosis.
Collapse
Affiliation(s)
- Yujie Shi
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.,School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Wenxiang Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.,School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yinlong Cheng
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.,School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Chang Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.,School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Siyu Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.,School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| |
Collapse
|
31
|
Riera-Heredia N, Lutfi E, Gutiérrez J, Navarro I, Capilla E. Fatty acids from fish or vegetable oils promote the adipogenic fate of mesenchymal stem cells derived from gilthead sea bream bone potentially through different pathways. PLoS One 2019; 14:e0215926. [PMID: 31017945 PMCID: PMC6481918 DOI: 10.1371/journal.pone.0215926] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 04/10/2019] [Indexed: 01/01/2023] Open
Abstract
Fish are rich in n-3 long-chain polyunsaturated fatty acids (LC-PUFA), such as eicosapentaenoic (EPA) and docosahexaenoic (DHA) acids, thus they have a great nutritional value for human health. In this study, the adipogenic potential of fatty acids commonly found in fish oil (EPA and DHA) and vegetable oils (linoleic (LA) and alpha-linolenic (ALA) acids), was evaluated in bone-derived mesenchymal stem cells (MSCs) from gilthead sea bream. At a morphological level, cells adopted a round shape upon all treatments, losing their fibroblastic form and increasing lipid accumulation, especially in the presence of the n-6 PUFA, LA. The mRNA levels of the key transcription factor of osteogenesis, runx2 significantly diminished and those of relevant osteogenic genes remained stable after incubation with all fatty acids, suggesting that the osteogenic process might be compromised. On the other hand, transcript levels of the main adipogenesis-inducer factor, pparg increased in response to EPA. Nevertheless, the specific PPARγ antagonist T0070907 appeared to suppress the effects being caused by EPA over adipogenesis. Moreover, LA, ALA and their combinations, significantly up-regulated the fatty acid transporter and binding protein, fatp1 and fabp11, supporting the elevated lipid content found in the cells treated with those fatty acids. Overall, this study has demonstrated that fatty acids favor lipid storage in gilthead sea bream bone-derived MSCs inducing their fate into the adipogenic versus the osteogenic lineage. This process seems to be promoted via different pathways depending on the fatty acid source, being vegetable oils-derived fatty acids more prone to induce unhealthier metabolic phenotypes.
Collapse
Affiliation(s)
- Natàlia Riera-Heredia
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Esmail Lutfi
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Joaquim Gutiérrez
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Isabel Navarro
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Encarnación Capilla
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
- * E-mail:
| |
Collapse
|
32
|
Ishaq A, Dufour D, Cameron K, von Zglinicki T, Saretzki G. Metabolic memory of dietary restriction ameliorates DNA damage and adipocyte size in mouse visceral adipose tissue. Exp Gerontol 2018; 113:228-236. [PMID: 30312736 DOI: 10.1016/j.exger.2018.10.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 09/24/2018] [Accepted: 10/08/2018] [Indexed: 11/30/2022]
Abstract
Dietary restriction (DR) is thought to exert its beneficial effects on healthspan at least partially by a senolytic and senostatic action, i.e. by reducing frequencies of cells with markers of DNA damage and senescence in multiple tissues. Due to its importance in metabolic and inflammation regulation, fat is a prime tissue for health span determination as well as a prime target for DR. We aimed to determine here whether the beneficial effects of DR would be retained over a subsequent period of ad libitum (AL) feeding. Male mice were kept under either 40% DR or AL feeding regimes from 3 to 12 months of age and then either switched back to the opposite feeding regimen or kept in the same state for another 3 months. Visceral adipose tissue from 4 to 5 mice per group for all conditions was analysed for markers of senescence (adipocyte size, γH2A.X, p16, p21) and inflammation (e.g. IL-6, TNFα, IL-1β) using immuno-staining or qPCR. Macrophages were detected by immunohistochemistry. We found that both 9 and 12 months DR (long term) as well as 3 month (short term, mid-life onset) DR reduced the number of cells harbouring DNA damage and adipocyte size (area and perimeter) in visceral adipocytes with similar efficiency. Importantly, beneficial health markers induced by DR such as small adipocyte size and low DNA damage were maintained for at least 3 month after termination of DR, demonstrating that the previously identified 'metabolic memory' of the DR state in male mice extends to senescence markers in visceral fat.
Collapse
Affiliation(s)
- Abbas Ishaq
- The Ageing Biology Centre, Newcastle Institute for Ageing, Institute for Cell and Molecular Biosciences, Campus of Ageing and Vitality, Newcastle upon Tyne, UK
| | - Damien Dufour
- The Ageing Biology Centre, Newcastle Institute for Ageing, Institute for Cell and Molecular Biosciences, Campus of Ageing and Vitality, Newcastle upon Tyne, UK
| | - Kerry Cameron
- The Ageing Biology Centre, Newcastle Institute for Ageing, Institute for Cell and Molecular Biosciences, Campus of Ageing and Vitality, Newcastle upon Tyne, UK
| | - Thomas von Zglinicki
- The Ageing Biology Centre, Newcastle Institute for Ageing, Institute for Cell and Molecular Biosciences, Campus of Ageing and Vitality, Newcastle upon Tyne, UK
| | - Gabriele Saretzki
- The Ageing Biology Centre, Newcastle Institute for Ageing, Institute for Cell and Molecular Biosciences, Campus of Ageing and Vitality, Newcastle upon Tyne, UK.
| |
Collapse
|
33
|
Kühn G, Pallauf K, Schulz C, Rimbach G. Flavonoids as putative modulators of Δ4-, Δ5-, and Δ6-desaturases: Studies in cultured hepatocytes, myocytes, and adipocytes. Biofactors 2018; 44:485-495. [PMID: 30365230 DOI: 10.1002/biof.1443] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 06/07/2018] [Accepted: 06/27/2018] [Indexed: 12/19/2022]
Abstract
This study was conducted to screen flavonoids for affecting expression of desaturases involved in omega-3 fatty acid synthesis and ceramide (CER) metabolism. To this end, cultured HepG2 hepatocytes, C2C12 myocytes, and 3T3-L1 adipocytes were treated with nontoxic concentrations of 12 selected flavonoids and expression of Δ4-, Δ5-, and Δ6-desaturases (DEGS1, FADS1, and FADS2, respectively) was determined. The flavonoids tested were more cytotoxic to HepG2 and 3T3-L1 than to C2C12 cells. In HepG2 cells, FADS1 was induced by quercetin and FADS2 expression was increased by daidzein, genistein, and pratensein treatment. DEGS1 was increased by apigenin, luteolin, orobol, and quercetin administration. In differentiated C2C12 cells, substances had no inducing effect or even lowered target gene expression. Pratensein induced both FADS1 and FADS2 in differentiated 3T3-L1 cells and DEGS1 was increased by treatment with apigenin, genistein, luteolin, orobol, and quercetin. In conclusion, pratensein may be an interesting test compound for further studies in vitro and in vivo on omega-3 synthesis since it induces its rate-limiting enzyme FADS2. Apigenin, luteolin, orobol, and quercetin induced DEGS1 and thereby possibly synthesis of proapoptotic CER in malignant HepG2 cells and 3T3-L1. In contrast, in benign C2C12 cells, they did not elevate mRNA steady state levels of DEGS1. That may partly explain the higher resistance of C2C12 cells against flavonoids compared to the other cell lines. By affecting tumor cells and nontumor cells differently, these flavonoids may be promising substances for further research regarding anticancer properties. © 2018 BioFactors, 44(5):485-495, 2018.
Collapse
Affiliation(s)
- Gianna Kühn
- Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Kathrin Pallauf
- Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Carsten Schulz
- Institute of Animal Breeding and Husbandry, University of Kiel, Kiel, Germany
- GMA-Gesellschaft für Marine Aquakultur mbH, Büsum, Germany
| | - Gerald Rimbach
- Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| |
Collapse
|
34
|
Zafar U, Khaliq S, Ahmad HU, Manzoor S, Lone KP. Metabolic syndrome: an update on diagnostic criteria, pathogenesis, and genetic links. Hormones (Athens) 2018; 17:299-313. [PMID: 30171523 DOI: 10.1007/s42000-018-0051-3] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 06/19/2018] [Indexed: 02/06/2023]
Abstract
Metabolic syndrome (MetS), today a major global public health problem, is a cluster of clinical, metabolic, and biochemical abnormalities, such as central adiposity, hypertension, insulin resistance, and dyslipidemias. These MetS-related traits significantly increase the risk of type 2 diabetes mellitus, adverse cardiac events, stroke, and hepatic steatosis. The pathogenesis of MetS is multifactorial, with the interplay of environmental, nutritional, and genetic factors. Chronic low-grade inflammation together with visceral adipose tissue, adipocyte dysfunction, and insulin resistance plays a major role in the progression of the syndrome by impairing lipid and glucose homeostasis in insulin-sensitive tissues, such as the liver, muscle, and adipocytes. Adipose-derived inflammatory cytokines and non-esterified fatty acids establish the link between central obesity IR, inflammation, and atherogenesis. Various studies have reported an association between MetS and related traits with single-nucleotide polymorphisms of different susceptibility genes. Modulation of cytokine levels, pro-oxidants, and disturbed energy homeostasis, in relation to the genetic variations, is described in this review of the recent literature, which also provides updated data regarding the epidemiology, diagnostic criteria, and pathogenesis of MetS.
Collapse
Affiliation(s)
- Uzma Zafar
- Department of Physiology & Cell Biology, University of Health Sciences, Lahore, Pakistan.
- Department of Physiology, Lahore Medical and Dental College, Lahore, Pakistan.
| | - Saba Khaliq
- Department of Physiology & Cell Biology, University of Health Sciences, Lahore, Pakistan
| | - Hafiz Usman Ahmad
- Department of Physiology & Cell Biology, University of Health Sciences, Lahore, Pakistan
| | - Sobia Manzoor
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, NUST, Islamabad, Pakistan
| | - Khalid P Lone
- Department of Physiology & Cell Biology, University of Health Sciences, Lahore, Pakistan.
| |
Collapse
|
35
|
Jin W, Jin W, Pan D. Ifi27 is indispensable for mitochondrial function and browning in adipocytes. Biochem Biophys Res Commun 2018; 501:273-279. [PMID: 29730295 DOI: 10.1016/j.bbrc.2018.04.234] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 04/30/2018] [Indexed: 11/26/2022]
Abstract
Brown adipose tissue (BAT) is specialized for energy expenditure, but the signaling pathways that regulate BAT metabolism and activity are incompletely understood. Interferon (IFN) signaling is a sophisticated defense mechanism to counteract viral infection. IFNs and interferon-stimulated genes (ISGs) are reported to exert profound effects on adipocytes. IFN-α inducible protein 27 (Ifi27/ISG12a) is a BAT-enriched gene, yet no any studies on its roles in BAT have been reported. Here, we show that Ifi27 protein localizes to mitochondria and the expression of Ifi27 can be induced by β3-adrenergic activation in adipose tissues. Knockdown of Ifi27 leads to reduced expression of key enzymes of tricarboxylic acid cycle (TCA), the subunits of electron transport chain (ETC) and uncoupling protein 1 (Ucp1) in brown and beige adipocytes. Moreover, the browning of subcutaneous white fat induced by β3-adrenergic agonist is also dramatically blocked. Ectopic expression of Ifi27 in brown adipocytes has the opposite effects. Together, these data indicate that Ifi27 regulates mitochondrial function and browning in adipocytes.
Collapse
Affiliation(s)
- Weiwei Jin
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai, 200032, China
| | - Wenfang Jin
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai, 200032, China
| | - Dongning Pan
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai, 200032, China.
| |
Collapse
|
36
|
Liu C, Shen YJ, Tu QB, Zhao YR, Guo H, Wang J, Zhang L, Shi HW, Sun Y. Pedunculoside, a novel triterpene saponin extracted from Ilex rotunda, ameliorates high-fat diet induced hyperlipidemia in rats. Biomed Pharmacother 2018. [PMID: 29518607 DOI: 10.1016/j.biopha.2018.02.131] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Pedunculoside (PE) is a novel triterpene saponin extracted from the dried barks of Ilex rotunda Thunb. The present study aims to explore lipid-lowering effects of PE on hyperlipidemia rat induced by high-fat diet. The rats were fed with the high-fat diet and subjected to intragastric administration of PE at doses of 30, 15, or 5 mg/kg daily for 7 weeks. The results demonstrated that treatment with PE for 7-week dramatically decreased serum total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C) and reduced liver TC in hyperlipidemia rat induced by high-fat diet. Furthermore, the results also showed that PE modulated the expression of enzymes involved in lipid metabolism including peroxisome proliferator-activated receptor α (PPAR-α), sterol regulatory element-binding protein 1 (SREBP-1), fatty acid synthase (FAS) and stearoyl CoA desaturase-1 (SCD-1) mRNA in liver. Besides, PE-treated group decreased weights and diameters of epididymal adipose hyperlipidemia rat. Mechanism study demonstrated that PE regulated PPAR-γ, CCAAT/Enhancer-binding Protein α (C/EBPα)、and SREBP-1 expression as well as inhibited phosphorylation of AMPK in MDI (methylisobutylxanthine, dexamethasone, insulin) induced-3T3L1 cells. Molecular Docking confirmed interaction between PE with proteins involving PPAR-γ, C/EBPα and SREBP-1. In summary, these findings may support that PE is a novel lipid-lowering drug candidate.
Collapse
Affiliation(s)
- Chang Liu
- College of Medicine, Yangzhou University, Yangzhou 225001, Jiangsu, PR China; School of Pharmacy, University of Rhode Island, RI, 02881, United States
| | - Yan-Jun Shen
- College of Medicine, Yangzhou University, Yangzhou 225001, Jiangsu, PR China
| | - Qing-Bo Tu
- College of Hanlin, Nanjing University of China Medicine, Taizhou 225300, Jiangsu, PR China
| | - Yan-Ran Zhao
- College of Hanlin, Nanjing University of China Medicine, Taizhou 225300, Jiangsu, PR China
| | - Hao Guo
- School of Pharmacy, University of Rhode Island, RI, 02881, United States; Department of Dermatology, No. 1 Hospital of China Medical University, 155N. Nanjing Street, Shenyang 110001, PR China
| | - Juan Wang
- College of Medicine, Yangzhou University, Yangzhou 225001, Jiangsu, PR China; College of Hanlin, Nanjing University of China Medicine, Taizhou 225300, Jiangsu, PR China
| | - Li Zhang
- School of Pharmacy, University of Missouri-Kansas City, MO, 64108, United States
| | - Hua-Wei Shi
- College of Medicine, Yangzhou University, Yangzhou 225001, Jiangsu, PR China
| | - Yun Sun
- College of Medicine, Yangzhou University, Yangzhou 225001, Jiangsu, PR China; College of Hanlin, Nanjing University of China Medicine, Taizhou 225300, Jiangsu, PR China.
| |
Collapse
|
37
|
Namkoong S, Cho CS, Semple I, Lee JH. Autophagy Dysregulation and Obesity-Associated Pathologies. Mol Cells 2018; 41:3-10. [PMID: 29370691 PMCID: PMC5792710 DOI: 10.14348/molcells.2018.2213] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 10/02/2017] [Accepted: 10/10/2017] [Indexed: 12/20/2022] Open
Abstract
Autophagy is one of the major degradative mechanisms that can eliminate excessive nutrients, toxic protein aggregates, damaged organelles and invading microorganisms. In response to obesity and obesity-associated lipotoxic, proteotoxic and oxidative stresses, autophagy plays an essential role in maintaining physiological homeostasis. However, obesity and its associated stress insults can often interfere with the autophagic process through various mechanisms, which result in further aggravation of obesity-related metabolic pathologies in multiple metabolic organs. Paradoxically, inhibition of autophagy, within specific contexts, indirectly produces beneficial effects that can alleviate several detrimental consequences of obesity. In this minireview, we will provide a brief discussion about our current understanding of the impact of obesity on autophagy and the role of autophagy dysregulation in modulating obesity-associated pathological outcomes.
Collapse
Affiliation(s)
- Sim Namkoong
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109-2200,
USA
| | - Chun-Seok Cho
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109-2200,
USA
| | - Ian Semple
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109-2200,
USA
| | - Jun Hee Lee
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109-2200,
USA
- Institute of Gerontology, University of Michigan, Ann Arbor, Michigan 48109-2200,
USA
| |
Collapse
|
38
|
Alsabeeh N, Chausse B, Kakimoto PA, Kowaltowski AJ, Shirihai O. Cell culture models of fatty acid overload: Problems and solutions. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1863:143-151. [PMID: 29155055 DOI: 10.1016/j.bbalip.2017.11.006] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 11/09/2017] [Accepted: 11/14/2017] [Indexed: 12/17/2022]
Abstract
High plasma levels of fatty acids occur in a variety of metabolic diseases. Cellular effects of fatty acid overload resulting in negative cellular responses (lipotoxicity) are often studied in vitro, in an attempt to understand mechanisms involved in these diseases. Fatty acids are poorly soluble, and thus usually studied when complexed to albumins such as bovine serum albumin (BSA). The conjugation of fatty acids to albumin requires care pertaining to preparation of the solutions, effective free fatty acid concentrations, use of different fatty acid species, types of BSA, appropriate controls and ensuring cellular fatty acid uptake. This review discusses lipotoxicity models, the potential problems encountered when using these cellular models, as well as practical solutions for difficulties encountered.
Collapse
Affiliation(s)
- Nour Alsabeeh
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA; Department of Physiology and Biophysics, Boston University School of Medicine, Boston, MA 02118, USA; Department of Physiology, Faculty of Medicine, Kuwait University, Kuwait
| | - Bruno Chausse
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP 05508-000, Brazil
| | - Pamela A Kakimoto
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP 05508-000, Brazil
| | - Alicia J Kowaltowski
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP 05508-000, Brazil.
| | - Orian Shirihai
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| |
Collapse
|
39
|
Hadj Ahmed S, Kaoubaa N, Kharroubi W, Zarrouk A, Najjar MF, Batbout F, Gamra H, Lizard G, Hammami M. Association of plasma fatty acid alteration with the severity of coronary artery disease lesions in Tunisian patients. Lipids Health Dis 2017; 16:154. [PMID: 28806974 PMCID: PMC5557073 DOI: 10.1186/s12944-017-0538-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 08/02/2017] [Indexed: 11/12/2022] Open
Abstract
Background Some factors related to diet are known to be involved in the progression of atherosclerosis in humans. Methods The relationship between plasma fatty acid (FA) levels and the severity of coronary artery disease (CAD), evaluated by Gensini score (GS), was investigated in CAD Tunisian patients compared to controls. Lipid profiles were analyzed, GS was calculated in CAD and non-CAD patients and compared to controls. Results CAD patients showed an alteration of conventional lipid parameters. In fact, a significant increase of plasmatic triglycerides (TG) level, atherogenic lipid ratios (TC/HDL-C,TG/HDL-C, LDL-C/HDL-C); and ApoB/ApoA1 was observed in the CAD group comparatively to controls (p < 0.001). Gensini score was showed to be a good indicator to evaluate cholesterol metabolism disorders associated with HDL-C since a negative association was found between HDL-C levels and GS for the two groups of patients. In addition, in the relation with FA and classes of FA, a negative association was found as expected, between Gensini score and total MUFA, PUFA n-3, total PUFA, GLA, DGLA and DHA. Furthermore, a positive association with stearic and erucic acid was found. Suggests that, GS is also a good indicator to evaluate FA metabolic disorders. Higher elongation index and modifications of desaturation index (D5D, D6D and D9D) were observed in patients compared to controls, supporting FA metabolism modifications. Conclusions In conclusion, although that Tunisian population appears to follow the Mediterranean diet, variations of plasmatic FA levels and desaturase activities in CAD patients highlights an alteration of FA metabolism and suggests an important implication of certain FA in the development of atherosclerosis.
Collapse
Affiliation(s)
- Samia Hadj Ahmed
- Faculty of Medicine, Research Laboratory LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', University of Monastir (Tunisia), Avicene st, 5019, Monastir, Tunisia.
| | - Nadia Kaoubaa
- Faculty of Medicine, Research Laboratory LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', University of Monastir (Tunisia), Avicene st, 5019, Monastir, Tunisia
| | - Wafa Kharroubi
- Faculty of Medicine, Research Laboratory LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', University of Monastir (Tunisia), Avicene st, 5019, Monastir, Tunisia
| | - Amira Zarrouk
- Faculty of Medicine, Research Laboratory LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', University of Monastir (Tunisia), Avicene st, 5019, Monastir, Tunisia
| | | | - Fathi Batbout
- Cardiology Department CHU Fattouma Bourguiba Monastir- Tunisia, Monastir, Tunisia
| | - Habib Gamra
- Cardiology Department CHU Fattouma Bourguiba Monastir- Tunisia, Monastir, Tunisia
| | - Gerard Lizard
- Team 'Biochemistry of Peroxisome, Inflammation and Lipid Metabolism' EA 7270 /University of Bourgogne-Franche Comté / INSERM, Dijon, France
| | - Mohamed Hammami
- Faculty of Medicine, Research Laboratory LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', University of Monastir (Tunisia), Avicene st, 5019, Monastir, Tunisia
| |
Collapse
|
40
|
Fat-enriched rather than high-fructose diets promote whitening of adipose tissue in a sex-dependent manner. J Nutr Biochem 2017; 49:22-29. [PMID: 28863366 DOI: 10.1016/j.jnutbio.2017.07.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 06/25/2017] [Accepted: 07/19/2017] [Indexed: 12/12/2022]
Abstract
Adipose tissue is a critical regulator of energy metabolism and an effector organ of excessive caloric intake. We studied the effects of high-fructose (HFruD), high-fat (HFD) and mixed high-sucrose and high-fat diet (HFHSD) on adipocyte morphology and biology and consecutive metabolic effects in male and female C57BL/6 mice. Forty male and 40 female mice were randomly assigned to one of four dietary groups and fed for 10 weeks ad libitum. After 10 weeks of feeding, mice were analyzed in regard to glucose metabolism, insulin sensitivity and alteration in adipocyte morphology and function. Weight gain and diminished insulin sensitivity in HFD- and HFHSD-fed mice were accompanied by increased adipocyte size and a shift in size distribution towards larger adipocytes in all mice. The latter effect was also found but less pronounced in HFruD-fed mice, while insulin sensitivity and body weight remained unaffected. In male mice, expansion of white adipocytes along with decreased uncoupling protein 1 (UCP-1) expression and alterations of mitochondrial biogenesis was found after HFD and HFHSD feeding, while in female mice, UCP-1 expression was also reduced in the HFruD dietary group. Diet-induced cellular alterations were less pronounced in female mice. Our data demonstrate that high-fat rather than high fructose consumption drives metabolically disadvantageous alterations of adipocyte differentiation involving whitening and insulin resistance in a sex-dependent manner with most deleterious effects seen upon administration of combined sucrose and fat-enriched diet in male mice.
Collapse
|
41
|
Selen Alpergin ES, Bolandnazar Z, Sabatini M, Rogowski M, Chiellini G, Zucchi R, Assadi-Porter FM. Metabolic profiling reveals reprogramming of lipid metabolic pathways in treatment of polycystic ovary syndrome with 3-iodothyronamine. Physiol Rep 2017; 5:e13097. [PMID: 28082426 PMCID: PMC5256158 DOI: 10.14814/phy2.13097] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 11/27/2016] [Accepted: 11/29/2016] [Indexed: 01/16/2023] Open
Abstract
Complex diseases such as polycystic ovary syndrome (PCOS) are associated with intricate pathophysiological, hormonal, and metabolic feedbacks that make their early diagnosis challenging, thus increasing the prevalence risks for obesity, cardiovascular, and fatty liver diseases. To explore the crosstalk between endocrine and lipid metabolic pathways, we administered 3-iodothyronamine (T1AM), a natural analog of thyroid hormone, in a mouse model of PCOS and analyzed plasma and tissue extracts using multidisciplinary omics and biochemical approaches. T1AM administration induces a profound tissue-specific antilipogenic effect in liver and muscle by lowering gene expression of key regulators of lipid metabolism, PTP1B and PLIN2, significantly increasing metabolites (glucogenic, amino acids, carnitine, and citrate) levels, while enhancing protection against oxidative stress. In contrast, T1AM has an opposing effect on the regulation of estrogenic pathways in the ovary by upregulating STAR, CYP11A1, and CYP17A1. Biochemical measurements provide further evidence of significant reduction in liver cholesterol and triglycerides in post-T1AM treatment. Our results shed light onto tissue-specific metabolic vs. hormonal pathway interactions, thus illuminating the intricacies within the pathophysiology of PCOS This study opens up new avenues to design drugs for targeted therapeutics to improve quality of life in complex metabolic diseases.
Collapse
Affiliation(s)
- Ebru S Selen Alpergin
- Department of Biological Chemistry, Johns Hopkins University, Baltimore, Maryland
- Department of Zoology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Zeinab Bolandnazar
- Department of Zoology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Martina Sabatini
- Dipartimento di Patologia Chirurgica, Medica, Molecolare e Area Critica, Università di Pisa, Pisa, Italy
| | - Michael Rogowski
- Department of Medicine, University of Alabama Birmingham, Birmingham, Alabama
| | - Grazia Chiellini
- Dipartimento di Patologia Chirurgica, Medica, Molecolare e Area Critica, Università di Pisa, Pisa, Italy
| | - Riccardo Zucchi
- Dipartimento di Patologia Chirurgica, Medica, Molecolare e Area Critica, Università di Pisa, Pisa, Italy
| | - Fariba M Assadi-Porter
- Department of Zoology, University of Wisconsin-Madison, Madison, Wisconsin
- Magnetic Resonance Facility at Madison, Madison, Wisconsin
| |
Collapse
|
42
|
Genetic Manipulation with Viral Vectors to Assess Metabolism and Adipose Tissue Function. Methods Mol Biol 2017; 1566:109-124. [PMID: 28244045 DOI: 10.1007/978-1-4939-6820-6_11] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Viral vectors have become widely used tools for genetic manipulation of adipose tissues to understand the biology and function of adipocytes in metabolism. There are a number of different viral vectors commonly used: retrovirus, lentivirus, adenovirus, and adeno-associated virus (AAV). Here, we review examples from the literature and describe methods to transduce adipocytes and adipose tissues using retrovirus, lentivirus, adenovirus, and AAV to ascertain gene function in adipose biology.
Collapse
|
43
|
de Carvalho PM, Gavião MBD, Carpenter GH. Altered autophagy and sympathetic innervation in salivary glands from high-fat diet mice. Arch Oral Biol 2016; 75:107-113. [PMID: 27825677 DOI: 10.1016/j.archoralbio.2016.10.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 10/01/2016] [Accepted: 10/25/2016] [Indexed: 12/21/2022]
Abstract
OBJECTIVE to investigate the effects of a high fat diet (HFD) on salivary glands in vivo, in a mouse model. In particular, whether it will induce the appearance of fat cells in salivary glands, alterations related to autophagy, mTOR pathway and sympathetic innervation. DESIGN 27 adult female ICR mice were separated in six groups. Three groups fed with (HFD) containing 55% fat, for one, two and three month and another three groups fed with normal diet (2.7% of fat), for the same time periods. The submandibular glands and liver were dissected and part homogenized for protein analyses and part fixed in formalin for histological analyses. RESULTS After three months the HFD fed mice total body weight fold change increased compared to controls. The Oil Red O staining showed no fat cells deposit in salivary gland however a large increase was observed in liver after three months of HFD. Adiponectin levels were significantly decreased in the HFD group after three months. The group fed with HFD for three months showed increased conversion of the LC3 autophagy marker in salivary gland. mTOR showed no activation regarding the time point studied. Tyrosine hydroxylase significantly decreased after two and three month of HFD. CONCLUSION HFD caused several changes after three months however the earliest change was noticed after two months regarding sympathetic innervation. This suggests neural alteration may drive other diet induced changes in salivary glands. These early changes may be the starting point for longer term alterations of salivary glands with alterations in diet.
Collapse
Affiliation(s)
- Polliane Morais de Carvalho
- Salivary Research, Department of Mucosal and Salivary Biology, King's College London Dental Institute, London, SE1 9RT, United Kingdom.
| | - Maria Beatriz Duarte Gavião
- Department of Pediatric Dentistry, Piracicaba Dental School,University of Campinas (UNICAMP), Av Limeira 901, Piracicaba, SP, 13414-903, Brazil
| | - Guy Howard Carpenter
- Salivary Research, Department of Mucosal and Salivary Biology, King's College London Dental Institute, London, SE1 9RT, United Kingdom
| |
Collapse
|
44
|
Cook KL, Soto-Pantoja DR, Clarke PAG, Cruz MI, Zwart A, Wärri A, Hilakivi-Clarke L, Roberts DD, Clarke R. Endoplasmic Reticulum Stress Protein GRP78 Modulates Lipid Metabolism to Control Drug Sensitivity and Antitumor Immunity in Breast Cancer. Cancer Res 2016; 76:5657-5670. [PMID: 27698188 PMCID: PMC5117832 DOI: 10.1158/0008-5472.can-15-2616] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 06/03/2016] [Indexed: 02/07/2023]
Abstract
The unfolded protein response is an endoplasmic reticulum stress pathway mediated by the protein chaperone glucose regulated-protein 78 (GRP78). Metabolic analysis of breast cancer cells shows that GRP78 silencing increases the intracellular concentrations of essential polyunsaturated fats, including linoleic acid. Accumulation of fatty acids is due to an inhibition of mitochondrial fatty acid transport, resulting in a reduction of fatty acid oxidation. These data suggest a novel role of GRP78-mediating cellular metabolism. We validated the effect of GRP78-regulated metabolite changes by treating tumor-bearing mice with tamoxifen and/or linoleic acid. Tumors treated with linoleic acid plus tamoxifen exhibited reduced tumor area and tumor weight. Inhibition of either GRP78 or linoleic acid treatment increased MCP-1 serum levels, decreased CD47 expression, and increased macrophage infiltration, suggesting a novel role for GRP78 in regulating innate immunity. GRP78 control of fatty acid oxidation may represent a new homeostatic function for GRP78. Cancer Res; 76(19); 5657-70. ©2016 AACR.
Collapse
Affiliation(s)
- Katherine L Cook
- Department of Surgery and Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina. Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC.
| | - David R Soto-Pantoja
- Department of Surgery and Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Pamela A G Clarke
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - M Idalia Cruz
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - Alan Zwart
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - Anni Wärri
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - Leena Hilakivi-Clarke
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - David D Roberts
- Laboratory of Pathology, National Cancer Institute, NIH, Bethesda, Maryland
| | - Robert Clarke
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| |
Collapse
|
45
|
Karagozian R, Bhardwaj G, Wakefield DB, Baffy G. Obesity paradox in advanced liver disease: obesity is associated with lower mortality in hospitalized patients with cirrhosis. Liver Int 2016; 36:1450-6. [PMID: 27037497 DOI: 10.1111/liv.13137] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 03/24/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS To investigate how obesity impacts inpatient mortality, length of stay (LOS) and costs in patients with cirrhosis. Obesity is a growing epidemic associated with multiple co-morbidities, increased morbidity, and a significant economic burden on healthcare. Despite the overall harmful impact of obesity, the 'obesity paradox' has been described as decreased mortality among obese vs non-obese patients in various chronic medical conditions. METHODS Analysis of the Nationwide Inpatient Sample (NIS) for 2012, which contains data from 44 states and 4378 hospitals. Data from all cases with primary, secondary or tertiary discharge diagnosis of cirrhosis identified by International Classification of Diseases-9 code 571.2, 571.5 571.6 were included. Primary outcomes included inpatient mortality, LOS, and hospital charges. Obesity as a predictor of mortality was defined by a predetermined obesity co-morbidity variable. RESULTS A total of 32,605 patients were included. Crude mortality was lower for obese cirrhotic patients (2.7% vs 3.5%, P = 0.02) than for non-obese cirrhotic patients. In contrast, median LOS was longer (4 vs 3 days, P < 0.001) and median hospital charges were higher for obese cirrhotic patients ($26 803 vs $23 447, P < 0.001) In multivariate logistic regression, obesity was associated with a lower risk of inpatient mortality (OR=0.73, 95%CI: 0.55-0.95, P = 0.02). CONCLUSIONS In the acute care setting, obese patients with cirrhosis have lower mortality than non-obese patients with cirrhosis, longer hospitalizations and higher healthcare cost, providing new evidence for the obesity paradox in cirrhosis. Obese cirrhotic patients are more likely to have enhanced nutritional reserve which may play a role in survival during acute illness.
Collapse
Affiliation(s)
- Raffi Karagozian
- School of Medicine, University of Connecticut, Farmington, CT, USA.
| | - Gaurav Bhardwaj
- School of Medicine, University of Connecticut, Farmington, CT, USA
| | - Dorothy B Wakefield
- Center for Public Health and Health Policy, UConn Health, Farmington, CT, USA
| | - Gyorgy Baffy
- Division of Gastroenterology & Hepatology, VA Boston Healthcare System, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
46
|
Schindler C. Review: The metabolic syndrome as an endocrine disease: is there an effective pharmacotherapeutic strategy optimally targeting the pathogenesis? Ther Adv Cardiovasc Dis 2016; 1:7-26. [DOI: 10.1177/1753944707082662] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The metabolic syndrome (MetS) represents a combination of cardiovascular risk determinants such as obesity, insulin resistance and lipid abnormalities such as hypertriglyceridemia, increased free fatty acids, low high-density-cholesterol and hypertension. As a multiple component condition it imparts a doubling of relative risk for atherosclerotic cardiovascular disease (ASCVD). It is currently controversial which component of the syndrome carries what weight. There is even a considerable debate whether the risk for ASCVD is greater in patients diagnosed with MetS than that by the individual risk factors. At present, no unifying pathogenetic mechanism can explain the metabolic syndrome and there is no unique treatment for it. This review summarizes and critically reviews the currently available clinical and scientific evidence for the concept that the MetS is causally an endocrine disease and discusses pharmacotherapeutic strategies targeting the pathogenesis rather than single symptoms of the cluster.
Collapse
Affiliation(s)
- Christoph Schindler
- Institute of Clinical Pharmacology, Medical Faculty, Technical University of Dresden, Fiedlerstrasse 27, 01307 Dresden, Germany christoph.schindler@ tu-dresden.de
| |
Collapse
|
47
|
Bruce KD, Szczepankiewicz D, Sihota KK, Ravindraanandan M, Thomas H, Lillycrop KA, Burdge GC, Hanson MA, Byrne CD, Cagampang FR. Altered cellular redox status, sirtuin abundance and clock gene expression in a mouse model of developmentally primed NASH. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:584-93. [PMID: 27040510 PMCID: PMC4874946 DOI: 10.1016/j.bbalip.2016.03.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 03/07/2016] [Accepted: 03/25/2016] [Indexed: 02/06/2023]
Abstract
Background We have previously shown that high fat (HF) feeding during pregnancy primes the development of non-alcoholic steatohepatits (NASH) in the adult offspring. However, the underlying mechanisms are unclear. Aims Since the endogenous molecular clock can regulate hepatic lipid metabolism, we investigated whether exposure to a HF diet during development could alter hepatic clock gene expression and contribute to NASH onset in later life. Methods Female mice were fed either a control (C, 7% kcal fat) or HF (45% kcal fat) diet. Offspring were fed either a C or HF diet resulting in four offspring groups: C/C, C/HF, HF/C and HF/HF. NAFLD progression, cellular redox status, sirtuin expression (Sirt1, Sirt3), and the expression of core clock genes (Clock, Bmal1, Per2, Cry2) and clock-controlled genes involved in lipid metabolism (Rev-Erbα, Rev-Erbβ, RORα, and Srebp1c) were measured in offspring livers. Results Offspring fed a HF diet developed NAFLD. However HF fed offspring of mothers fed a HF diet developed NASH, coupled with significantly reduced NAD+/NADH (p < 0.05, HF/HF vs C/C), Sirt1 (p < 0.001, HF/HF vs C/C), Sirt3 (p < 0.01, HF/HF vs C/C), perturbed clock gene expression, and elevated expression of genes involved lipid metabolism, such as Srebp1c (p < 0.05, C/HF and HF/HF vs C/C). Conclusion Our results suggest that exposure to excess dietary fat during early and post-natal life increases the susceptibility to develop NASH in adulthood, involving altered cellular redox status, reduced sirtuin abundance, and desynchronized clock gene expression. Offspring of mothers fed a high fat diet show severe fatty liver in later life. HF feeding is associated with altered cellular redox status and reduced sirtuin gene expression. HF feeding desynchronises the expression of core clock genes and lipogenic transcription factors. Exposure to a HF diet during development causes changes in liver metabolism that precede severe fatty liver disease.
Collapse
Affiliation(s)
- Kimberley D Bruce
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK; University of Colorado Anschutz Medical Campus, Endocrinology, Metabolism and Diabetes, Aurora, USA.
| | - Dawid Szczepankiewicz
- Poznań University of Life Sciences, Department of Animal Physiology and Biochemistry, Poznań, Poland
| | - Kiran K Sihota
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Manoj Ravindraanandan
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Hugh Thomas
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Karen A Lillycrop
- Centre for Biological Sciences, Institute of Developmental Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton, UK
| | - Graham C Burdge
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Mark A Hanson
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Christopher D Byrne
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Felino R Cagampang
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| |
Collapse
|
48
|
Luo H, Zhou Y, Hu X, Peng X, Wei H, Peng J, Jiang S. Activation of PPARγ2 by PPARγ1 through a functional PPRE in transdifferentiation of myoblasts to adipocytes induced by EPA. Cell Cycle 2016; 14:1830-41. [PMID: 25892270 DOI: 10.1080/15384101.2015.1033594] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
PPARγ and Wnt signaling are central positive and negative regulators of adipogenesis, respectively. Here we identified that, eicosapentaenoic acid (EPA) could effectively induce the transdifferentiation of myoblasts into adipocytes through modulation of both PPARγ expression and Wnt signaling. During the early stage of transdifferentiation, EPA activates PPARδ and PPARγ1, which in turn targets β-catenin to degradation and down-regulates Wnt/β-catenin signaling, such that the myogenic fate of myoblasts could be switched to adipogenesis. In addition, EPA up-regulates the expression of PPARγ1 by activating RXRα, then PPARγ1 binds to the functional peroxisome proliferator responsive element (PPRE) in the promoter of adipocyte-specific PPARγ2 to continuously activate the expression of PPARγ2 throughout the transdifferentiation process. Our data indicated that EPA acts as a dual-function stimulator of adipogenesis that both inhibits Wnt signaling and induces PPARγ2 expression to facilitate the transdifferentiation program, and the transcriptional activation of PPARγ2 by PPARγ1 is not only the key factor for the transdifferentiation of myoblasts to adipocytes, but also the crucial evidence for successful transdifferentiation. The present findings provided insight for the first time as to how EPA induces the transdifferentiation of myoblasts to adipocytes, but also provide new clues for strategies to prevent and treat some metabolic diseases.
Collapse
Key Words
- BSA, bovine serum albumin
- C/EBP, CCAAT/enhancer-binding protein
- DHA, docosahexaenoic acid
- DMEM, Dulbecco's modified Eagle's medium
- EPA, eicosapentaenoic acid
- IMF, intramuscular fat
- PPAR, peroxisome proliferator-activated receptor
- PPARγ1
- PPARγ2
- PPARδ
- PPRE, peroxisome proliferator responsive element
- PUFA, polyunsaturated fatty acids
- RXR, retinoid X receptor.
- Wnt/β-catenin signaling
- eicosapentaenoic acid
- transdifferentiation
Collapse
Affiliation(s)
- Hefeng Luo
- a Department of Animal Nutrition and Feed Science; College of Animal Science and Technology; Huazhong Agricultural University ; Wuhan , China
| | | | | | | | | | | | | |
Collapse
|
49
|
Black PN, Ahowesso C, Montefusco D, Saini N, DiRusso CC. Fatty Acid Transport Proteins: Targeting FATP2 as a Gatekeeper Involved in the Transport of Exogenous Fatty Acids. MEDCHEMCOMM 2016; 7:612-622. [PMID: 27446528 DOI: 10.1039/c6md00043f] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The fatty acid transport proteins (FATP) are classified as members of the Solute Carrier 27 (Slc27) family of proteins based on their ability to function in the transport of exogenous fatty acids. These proteins, when localized to the plasma membrane or at intracellular membrane junctions with the endoplasmic reticulum, function as a gate in the regulated transport of fatty acids and thus represent a therapeutic target to delimit the acquisition of fatty acids that contribute to disease as in the case of fatty acid overload. To date, FATP1, FATP2, and FATP4 have been used as targets in the selection of small molecule inhibitors with the goal of treating insulin resistance and attenuating dietary absorption of fatty acids. Several studies targeting FATP1 and FATP4 were based on the intrinsic acyl CoA synthetase activity of these proteins and not on transport directly. While several classes of compounds were identified as potential inhibitors of fatty acid transport, in vivo studies using a mouse model failed to provide evidence these compounds were effective in blocking or attenuating fatty acid transport. Studies targeting FATP2 employed a naturally occurring splice variant, FATP2b, which lacks intrinsic acyl CoA synthetase due to the deletion of exon 3, yet is fully functional in fatty acid transport. These studies identified two compounds, 5'-bromo-5-phenyl-spiro[3H-1,3,4-thiadiazole-2,3'-indoline]-2'-one), now referred to as Lipofermata, and 2-benzyl-3-(4-chlorophenyl)-5-(4-nitrophenyl)pyrazolo[1,5-a]pyrimidin-7(4H)-one, now called Grassofermata, that are effective fatty acid transport inhibitors both in vitro using a series of model cell lines and in vivo using a mouse model.
Collapse
Affiliation(s)
- Paul N Black
- Department of Biochemistry, University of Nebraska, Lincoln, NE
| | | | | | - Nipun Saini
- Department of Biochemistry, University of Nebraska, Lincoln, NE
| | | |
Collapse
|
50
|
Jia T, Tynelius P, Rasmussen F. U-shaped association of body mass index in early adulthood with unintentional mortality from injuries: a cohort study of Swedish men with 35 years of follow-up. Int J Obes (Lond) 2015; 40:809-14. [PMID: 26607037 DOI: 10.1038/ijo.2015.239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 11/04/2015] [Accepted: 11/06/2015] [Indexed: 01/09/2023]
Abstract
OBJECTIVES To investigate the dose-response association between body mass index (BMI) in young adulthood and the risk of mortality caused by unintentional injuries. METHODS We performed a cohort study including 7 43 398 men identified by linkage of the Multigeneration Register and the Military Service Conscription Register. Cox regression models were used to examine crude and adjusted hazard ratios (HRs) and 95% confidence intervals (95% CIs) of the relationships between BMI at age 18-20 years and the risk of death from all unintentional injuries as well as from specific unintentional injuries. We then estimated the population attributable fractions (PAFs)-the proportion of unintentional deaths that was attributable to underweight, overweight and obesity in this population-based cohort. RESULTS During 35.9 years of follow-up, 6461 deaths occurred from unintentional injuries, including 3064 deaths from road injury, 978 from poisoning, 503 from falls, 243 from fire and 348 from drowning. Underweight subjects had a higher risk of mortality in all unintentional injuries (HR, 1.05; 95% CI, 1.03-1.10) and mortality in burns (HR, 1.65; 95% CI, 1.13-2.40) compared with BMI between 18.5 and 22.5 kg m(-2) (reference group). BMI >25 kg m(-2) was associated with increased risk of death from all unintentional injuries (HR, 1.36; 95% CI, 1.12-1.65) and road accidents (HR, 1.50; 95% CI, 1.14-1.97). Estimates of PAF suggested that 4.4% of the mortality in Swedish men caused by unintentional injuries could have been avoided if BMI values were kept between 18.5 and 22.5 kg m(-2). CONCLUSIONS A U-shaped association was observed between BMI and risk of unintentional death. Both underweight and overweight were associated with increased mortality risk for all unintentional injuries and for subtype causes. Our study suggests that BMI might be a significant target for preventive interventions on deaths caused by unintentional injuries.
Collapse
Affiliation(s)
- T Jia
- Child and Adolescent Public Health Epidemiology Unit, Department of Public Health Sciences, Karolinska Institutet, Stockholm, Sweden
| | - P Tynelius
- Child and Adolescent Public Health Epidemiology Unit, Department of Public Health Sciences, Karolinska Institutet, Stockholm, Sweden
| | - F Rasmussen
- Child and Adolescent Public Health Epidemiology Unit, Department of Public Health Sciences, Karolinska Institutet, Stockholm, Sweden.,Centre for Epidemiology and Community Medicine, Stockholm County Council, Health Care Services, Stockholm, Sweden
| |
Collapse
|