51
|
Tan L, Zhang Y, Crowe-White KM, Senkus KE, Erwin ME, Wang H. Vitamin A Supplementation during Suckling and Postweaning Periods Attenuates the Adverse Metabolic Effects of Maternal High-Fat Diet Consumption in Sprague-Dawley Rats. Curr Dev Nutr 2020; 4:nzaa111. [PMID: 32734136 PMCID: PMC7382617 DOI: 10.1093/cdn/nzaa111] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/08/2020] [Accepted: 06/23/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Vitamin A (VA) has been demonstrated to be a regulator of adipose tissue (AT) development in adult obese models. However, little is known about the effect of VA on obesity-associated developmental and metabolic conditions in early life. OBJECTIVES We aimed to assess the effects of dietary VA supplementation during suckling and postweaning periods on the adiposity and metabolic health of neonatal and weanling rats from mothers consuming a high-fat diet (HFD). METHODS Pregnant Sprague-Dawley rats were fed a normal-fat diet (NFD; 25% fat; n = 2) or an HFD (50% fat; n = 2), both with 2.6 mg VA/kg. Upon delivery, half of the rat mothers were switched to diets with supplemented VA at 129 mg/kg, whereas the other half remained at 2.6 mg VA/kg. Four groups of rat pups were designated as NFD, NFD + VA, HFD, and HFD + VA, respectively. At postnatal day (P)14, P25, and P35, pups (n = 4 or 3/group) were killed. Body weight (BW), visceral white AT (WAT) mass, brown AT (BAT) mass, uncoupling protein 1 mRNA expression in BAT, serum glucose, lipids, adipokines, and inflammatory biomarkers, as well as serum and AT redox status were assessed. RESULTS Rat pups in the HFD group exhibited significantly higher BW, WAT mass, and serum glucose and leptin but reduced BAT mass compared with the NFD group. Without affecting the dietary intake, supplementing the HFD with VA significantly reduced the BW and WAT mass of pups but increased the BAT mass, significantly lowered the systemic and WAT oxidative stress, and modulated serum adipokines and lipids to some extent. CONCLUSIONS VA supplementation during suckling and postweaning periods attenuated metabolic perturbations caused by excessive fat intake. Supplementing maternal or infant obesogenic diets with VA or establishing a higher RDA of VA for specific populations should be studied further for managing overweight/obesity in early life.
Collapse
Affiliation(s)
- Libo Tan
- Department of Human Nutrition, University of Alabama, Tuscaloosa, AL, USA
| | - Yanqi Zhang
- Department of Human Nutrition, University of Alabama, Tuscaloosa, AL, USA
| | | | - Katelyn E Senkus
- Department of Human Nutrition, University of Alabama, Tuscaloosa, AL, USA
| | - Maddy E Erwin
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, USA
| | - Hui Wang
- Department of Human Nutrition, University of Alabama, Tuscaloosa, AL, USA
| |
Collapse
|
52
|
Zhang Y, Crowe-White KM, Kong L, Tan L. Vitamin A Status and Deposition in Neonatal and Weanling Rats Reared by Mothers Consuming Normal and High-Fat Diets with Adequate or Supplemented Vitamin A. Nutrients 2020; 12:nu12051460. [PMID: 32443575 PMCID: PMC7284941 DOI: 10.3390/nu12051460] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 12/13/2022] Open
Abstract
The circulating level of vitamin A (VA; retinol) was reported to be lower in obese adults. It is unknown if maternal obesity influences the VA status of offspring. The objective of the study was to determine the VA status and deposition of neonatal and weanling rats reared by mothers consuming a normal or high-fat diet (NFD or HFD) with or without supplemented VA. Pregnant Sprague-Dawley rats were randomized to an NFD or HFD with 2.6 mg/kg VA. Upon delivery, half of the rat mothers in the NFD or HFD cohort were switched to an NFD or HFD with supplemented VA at 129 mg/kg (NFD+VA and HFD+VA group). The other half remained on their original diet (NFD and HFD group). At postnatal day 14 (P14), P25, and P35, pups (n = 4 or 3/group/time) were euthanized. The total retinol concentration in the serum, liver, visceral white adipose tissue (WAT), and brown adipose tissue (BAT) was measured. At P14, the HFD+VA group showed a significantly lower serum VA than the NFD+VA group. At P25, both the VA concentration and total mass in the liver, WAT, and BAT were significantly higher in the HFD+VA than the NFD+VA group. At P35, the HFD group exhibited a significantly higher VA concentration and mass in the liver and BAT compared with the NFD group. In conclusion, maternal HFD consumption resulted in more VA accumulation in storage organs in neonatal and/or weanling rats, which potentially compromised the availability of VA in circulation, especially under the VA-supplemented condition.
Collapse
|
53
|
Kakoty V, K C S, Tang RD, Yang CH, Dubey SK, Taliyan R. Fibroblast growth factor 21 and autophagy: A complex interplay in Parkinson disease. Biomed Pharmacother 2020; 127:110145. [PMID: 32361164 DOI: 10.1016/j.biopha.2020.110145] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 03/27/2020] [Accepted: 04/04/2020] [Indexed: 12/22/2022] Open
Abstract
Parkinson disease (PD) is the second common neurodegenerative disorder after Alzheimer's disease (AD). The predominant pathological hallmark is progressive loss of dopaminergic (DA) neurones in the substantia nigra (SN) complicated by aggregation of misfolded forms of alpha-synuclein (α-syn). α-syn is a cytosolic synaptic protein localized in the presynaptic neuron under normal circumstances. What drives misfolding of this protein is largely unknown. However, recent studies suggest that autophagy might be an important risk factor for contributing towards PD. Autophagy is an evolutionarily conserved mechanism that causes the clearance or degradation of misfolded, mutated and damaged proteins, organelles etc. However, in an aging individual this process might deteriorate which could possibly lead to the accumulation of damaged proteins. Hence, autophagy modulation might provide some interesting cues for the treatment of PD. Additionally, Fibroblast growth factor 21 (FGF21) which is known for its role as a potent regulator of glucose and energy metabolism has also proved to be neuroprotective in various neurodegenerative conditions possibly via mediation of autophagy.
Collapse
Affiliation(s)
- Violina Kakoty
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, 333031, Rajasthan, India.
| | - Sarathlal K C
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, 333031, Rajasthan, India.
| | - Ruei-Dun Tang
- Department of Pharmacology, Taipei Medical University, Taipei, Taiwan.
| | - Chih Hao Yang
- Department of Pharmacology, Taipei Medical University, Taipei, Taiwan.
| | - Sunil Kumar Dubey
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, 333031, Rajasthan, India.
| | - Rajeev Taliyan
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, 333031, Rajasthan, India.
| |
Collapse
|
54
|
Xia Y, Feng H, Li ZW, Tang KX, Gao HQ, Wang WL, Cui XP, Li XL. Low-dose phloretin alleviates diabetic atherosclerosis through endothelial KLF2 restoration. Biosci Biotechnol Biochem 2020; 84:815-823. [PMID: 31791197 DOI: 10.1080/09168451.2019.1699396] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
ABSTRACT
We investigated whether low-dose phloretin served as daily dietary supplements could ameliorate diabetic atherosclerosis and the role of kruppel-like factor 2 (KLF2). HUVECs cultured in high glucose medium were treated with different concentrations of phloretin and KLF2 mRNA, and protein level was detected. Diabetes was induced using streptozotocin in Apoe−/- mice after which they were fed a high-cholesterol diet for 8 weeks. Diabetic mice injected with KLF2 shRNA-lentivirus or control virus were treated with 20 mg/kg phloretin. Glucose, lipid profile, aortic atheroma, and endothelial nitric oxide synthase (eNOS) expression were detected. Phloretin retained endothelial function by KLF2-eNOS activation under hyperglycemia. Low-dose phloretin helped with lipid metabolism, and blocked the acceleration of atherosclerosis in STZ-induced diabetic mice since the early stage, which was diminished by KLF2 knockdown. Low-dose phloretin exhibited athero-protective effect in diabetic Apoe−/- mice dependent on KLF2 activation. This finding makes phloretin for diabetic atherosclerosis.
Collapse
Affiliation(s)
- Yong Xia
- Department of Geriatric Medicine, Qi-lu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Hua Feng
- Department of Digestive Disease, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Zhen-Wei Li
- Department of Gastroenterology, Mengyin People’s Hospital, Mengyin, China
| | - Kuan-Xiao Tang
- Department of Geriatric Medicine, Qi-lu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Hai-Qing Gao
- Department of Geriatric Medicine, Qi-lu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Wei-Ling Wang
- Department of Geriatric Medicine, Qi-lu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Xiao-Pei Cui
- Department of Geriatric Medicine, Qi-lu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Xiao-Li Li
- Department of Drug Purchase and Supply, Qi-Lu Hospital of Shandong University, Jinan, China
| |
Collapse
|
55
|
Li Q, Zhang T, Zhang R, Qin X, Zhao J. All-trans retinoic acid regulates sheep primary myoblast proliferation and differentiation in vitro. Domest Anim Endocrinol 2020; 71:106394. [PMID: 31731254 DOI: 10.1016/j.domaniend.2019.106394] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 08/15/2019] [Accepted: 09/04/2019] [Indexed: 01/09/2023]
Abstract
Vitamin A and its metabolite, retinoic acid (RA), play key roles in cell differentiation and organ morphogenesis. The objective of this study was to investigate the effect of RA on sheep primary myoblast proliferation and differentiation. Sheep primary myoblasts were isolated and treated with all-trans retinoic acid (ATRA, 10 nM and 100 nM) and vehicle. The results showed that 10 nM ATRA sufficiently inhibited cell proliferation, which might be through downregulation of cyclin D1 (P < 0.05) and cyclin-dependent kinase 4 (P < 0.05) and proliferating cell nuclear antigen protein (P < 0.05) abundance. Moreover, compared with control cells, both 10 nM and 100 nM ATRA promoted myotube formation and increased fusion index (P < 0.05), which was associated with elevated myogenin mRNA content (P < 0.05). As expected, both myogenin (P < 0.01) and myosin heavy chain (P < 0.05) protein levels were increased by ATRA. Interestingly, ATRA treatment increased H3K4me3 and decreased H3K27me3 enrichment in the myogenin promoter region (P < 0.05). Meanwhile, 100 nM ATRA stimulated 2-(N-(7-Nitrobenz-2-oxa-1,3-diazol-4-yl) Amino)-2-deoxyglucose uptake (P < 0.05) and upregulated glucose transporter 4 expression at both mRNA and protein levels (P < 0.05). Although ATRA did not alter p38 content, phospho-p38 content was increased (P < 0.01). In addition, ATRA treatment activated the mTOR signaling pathway (P < 0.05). Taken together, these results demonstrated that ATRA plays an important role in regulating sheep myoblast proliferation and myogenic differentiation and suggested vitamin A as a potential target for manipulating muscle growth efficiency in sheep industry.
Collapse
Affiliation(s)
- Q Li
- Department of Animal Genetics & Breeding, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - T Zhang
- Department of Animal Genetics & Breeding, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - R Zhang
- Department of Animal Genetics & Breeding, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - X Qin
- Department of Animal Genetics & Breeding, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - J Zhao
- Department of Animal Genetics & Breeding, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China.
| |
Collapse
|
56
|
Abstract
Generation of the autacoid all-trans-retinoic acid (ATRA) from retinol (vitamin A) relies on a complex metabolon that includes retinol binding-proteins and enzymes from the short-chain dehydrogenase/reductase and aldehyde dehydrogenase gene families. Serum retinol binding-protein delivers all-trans-retinol (vitamin A) from blood to cells through two membrane receptors, Stra6 and Rbpr2. Stra6 and Rbpr2 convey retinol to cellular retinol binding-protein type 1 (Crbp1). Holo-Crbp1 delivers retinol to lecithin: retinol acyl transferase (Lrat) for esterification and storage. Lrat channels retinol directly into its active site from holo-Crbp1 by protein-protein interaction. The ratio apo-Crbp1/holo-Crbp1 directs flux of retinol into and out of retinyl esters, through regulating esterification vs ester hydrolysis. Multiple retinol dehydrogenases (Rdh1, Rdh10, Dhrs9, Rdhe2, Rdhe2s) channel retinol from holo-Crbp1 to generate retinal for ATRA biosynthesis. β-Carotene oxidase type 1 generates retinal from carotenoids, delivered by the scavenger receptor-B1. Retinal reductases (Dhrs3, Dhrs4, Rdh11) reduce retinal into retinol, thereby restraining ATRA biosynthesis. Retinal dehydrogenases (Raldh1, 2, 3) dehydrogenate retinal irreversibly into ATRA. ATRA regulates its own concentrations by inducing Lrat and ATRA degradative enzymes. ATRA exhibits hormesis. Its effects relate to its concentration as an inverted J-shaped curve, transitioning from beneficial in the "goldilocks" zone to toxicity, as concentrations increase. Hormesis has distorted understanding physiological effects of ATRA post-nataly using chow-diet fed, ATRA-dosed animal models. Cancer, immune deficiency and metabolic abnormalities result from mutations and/or insufficiency in Crbp1 and retinoid metabolizing enzymes.
Collapse
Affiliation(s)
- Joseph L Napoli
- Graduate Program in Metabolic Biology, Nutritional Sciences and Toxicology, University of California, Berkeley, CA, United States.
| |
Collapse
|
57
|
Rogers MA, Chen J, Nallamshetty S, Pham T, Goto S, Muehlschlegel JD, Libby P, Aikawa M, Aikawa E, Plutzky J. Retinoids Repress Human Cardiovascular Cell Calcification With Evidence for Distinct Selective Retinoid Modulator Effects. Arterioscler Thromb Vasc Biol 2020; 40:656-669. [PMID: 31852220 PMCID: PMC7047603 DOI: 10.1161/atvbaha.119.313366] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Retinoic acid (RA) is a ligand for nuclear receptors that modulate gene transcription and cell differentiation. Whether RA controls ectopic calcification in humans is unknown. We tested the hypothesis that RA regulates osteogenic differentiation of human arterial smooth muscle cells and aortic valvular interstitial cells that participate in atherosclerosis and heart valve disease, respectively. Approach and Results: Human cardiovascular tissue contains immunoreactive RAR (RA receptor)-a retinoid-activated nuclear receptor directing multiple transcriptional programs. RA stimulation suppressed primary human cardiovascular cell calcification while treatment with the RAR inhibitor AGN 193109 or RARα siRNA increased calcification. RA attenuated calcification in a coordinated manner, increasing levels of the calcification inhibitor MGP (matrix Gla protein) while decreasing calcification-promoting TNAP (tissue nonspecific alkaline phosphatase) activity. Given that nuclear receptor action varies as a function of distinct ligand structures, we compared calcification responses to cyclic retinoids and the acyclic retinoid peretinoin. Peretinoin suppressed human cardiovascular cell calcification without inducing either secretion of APOC3 (apolipoprotein-CIII), which promotes atherogenesis, or reducing CYP7A1 (cytochrome P450 family 7 subfamily A member 1) expression, which occurred with cyclic retinoids all-trans RA, 9-cis RA, and 13-cis RA. Additionally, peretinoin did not suppress human femur osteoblast mineralization, whereas all-trans RA inhibited osteoblast mineralization. CONCLUSIONS These results establish retinoid regulation of human cardiovascular calcification, provide new insight into mechanisms involved in these responses, and suggest selective retinoid modulators, like acyclic retinoids may allow for treating cardiovascular calcification without the adverse effects associated with cyclic retinoids.
Collapse
MESH Headings
- Alkaline Phosphatase
- Aortic Valve/drug effects
- Aortic Valve/metabolism
- Aortic Valve/pathology
- Apolipoprotein C-III/genetics
- Apolipoprotein C-III/metabolism
- Calcium-Binding Proteins/genetics
- Calcium-Binding Proteins/metabolism
- Carotid Arteries/drug effects
- Carotid Arteries/metabolism
- Carotid Arteries/pathology
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Cells, Cultured
- Cholesterol 7-alpha-Hydroxylase/genetics
- Cholesterol 7-alpha-Hydroxylase/metabolism
- Coronary Vessels/drug effects
- Coronary Vessels/metabolism
- Coronary Vessels/pathology
- Extracellular Matrix Proteins/genetics
- Extracellular Matrix Proteins/metabolism
- Heart Valve Diseases/genetics
- Heart Valve Diseases/metabolism
- Heart Valve Diseases/pathology
- Heart Valve Diseases/prevention & control
- Humans
- Isotretinoin/pharmacology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Osteogenesis/drug effects
- Receptors, Retinoic Acid/agonists
- Receptors, Retinoic Acid/genetics
- Receptors, Retinoic Acid/metabolism
- Retinoids/pharmacology
- Retinoids/toxicity
- Signal Transduction
- Tretinoin/pharmacology
- Vascular Calcification/genetics
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Vascular Calcification/prevention & control
- Matrix Gla Protein
Collapse
Affiliation(s)
- Maximillian A. Rogers
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| | - Jiaohua Chen
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| | - Shriram Nallamshetty
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| | - Tan Pham
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| | - Shinji Goto
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| | - Jochen D. Muehlschlegel
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| | - Peter Libby
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| | - Jorge Plutzky
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| |
Collapse
|
58
|
Bonet ML, Ribot J, Galmés S, Serra F, Palou A. Carotenoids and carotenoid conversion products in adipose tissue biology and obesity: Pre-clinical and human studies. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158676. [PMID: 32120014 DOI: 10.1016/j.bbalip.2020.158676] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 02/20/2020] [Accepted: 02/21/2020] [Indexed: 02/07/2023]
Abstract
Antiobesity activities of carotenoids and carotenoid conversion products (CCPs) have been demonstrated in pre-clinical studies, and mechanisms behind have begun to be unveiled, thus suggesting these compounds may help obesity prevention and management. The antiobesity action of carotenoids and CCPs can be traced to effects in multiple tissues, notably the adipose tissues. Key aspects of the biology of adipose tissues appear to be affected by carotenoid and CCPs, including adipogenesis, metabolic capacities for energy storage, release and inefficient oxidation, secretory function, and modulation of oxidative stress and inflammatory pathways. Here, we review the connections of carotenoids and CCPs with adipose tissue biology and obesity as revealed by cell and animal intervention studies, studies addressing the role of endogenous retinoid metabolism, and human epidemiological and intervention studies. We also consider human genetic variability influencing carotenoid and vitamin A metabolism, particularly in adipose tissues, as a potentially relevant aspect towards personalization of dietary recommendations to prevent or manage obesity and optimize metabolic health. This article is part of a Special Issue entitled Carotenoids recent advances in cell and molecular biology edited by Johannes von Lintig and Loredana Quadro.
Collapse
Affiliation(s)
- M Luisa Bonet
- Grup de Recerca Nutrigenòmica i Obesitat, Laboratori de Biologia Molecular, Nutrició i Biotecnologia (LBNB), Universitat de les Illes Balears, Palma de Mallorca, Spain; Institut d'Investigació Sanitària Illes Balears (IdISBa), Spain; CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), Spain.
| | - Joan Ribot
- Grup de Recerca Nutrigenòmica i Obesitat, Laboratori de Biologia Molecular, Nutrició i Biotecnologia (LBNB), Universitat de les Illes Balears, Palma de Mallorca, Spain; Institut d'Investigació Sanitària Illes Balears (IdISBa), Spain; CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), Spain
| | | | - Francisca Serra
- Grup de Recerca Nutrigenòmica i Obesitat, Laboratori de Biologia Molecular, Nutrició i Biotecnologia (LBNB), Universitat de les Illes Balears, Palma de Mallorca, Spain; Institut d'Investigació Sanitària Illes Balears (IdISBa), Spain; CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), Spain
| | - Andreu Palou
- Grup de Recerca Nutrigenòmica i Obesitat, Laboratori de Biologia Molecular, Nutrició i Biotecnologia (LBNB), Universitat de les Illes Balears, Palma de Mallorca, Spain; Institut d'Investigació Sanitària Illes Balears (IdISBa), Spain; CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), Spain
| |
Collapse
|
59
|
Xie L, Zou L, Chen J, Liu Y. All-Trans Retinoic Acid Inhibits Bone Marrow Mesenchymal Stem Cell Commitment to Adipocytes via Upregulating FRA1 Signaling. Int J Endocrinol 2020; 2020:6525787. [PMID: 32089684 PMCID: PMC7013307 DOI: 10.1155/2020/6525787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 12/25/2019] [Accepted: 01/07/2020] [Indexed: 11/23/2022] Open
Abstract
Obesity, caused by an increased number and volume of adipocytes, is a global epidemic that seriously threatens human health. Bone marrow mesenchymal stem cells (BMSCs) can differentiate into adipocytes. All-trans retinoic acid (atRA, the active form of vitamin A) inhibits the adipogenic differentiation of BMSCs through its receptor RARG. The expression level of FRA1 (FOS like 1, AP-1 transcription factor subunit) in atRA-treated BMSCs increased, suggesting that atRA-mediated inhibition of BMSCs adipogenesis involves FRA1. BMSCs were transfected with adenovirus overexpressing Fra1 (ad-fra1) or silenced for Fra1 (si-fra1) and then treated with atRA. BMSCs treated with atRA and treated with ad-fra1 showed decreased mRNA and protein levels of key adipogenic genes (Pparg2, Cebpa) and adipogenesis-associated genes (Cd36, Fabp, Lpl, and Plin); atRA had a stronger inhibitory effect on adipogenesis compared with that in the ad-fra1 group. Adipogenic gene expression in Fra1-silenced BMSCs was significantly upregulated. Compared with that in the atRA group, the si-fra1 + atRA also upregulated adipogenic gene expression. However, compared with si-fra1, si-fra1 + atRA significantly inhibited adipogenic differentiation. Chromatin immunoprecipitation showed that RARG directly regulates Fra1 and FRA1 directly regulates Pparg2 and Cebpa. The results supported the conclusion that atRA inhibits BMSC adipogenesis partially through the RARG-FRA1-PPARG2 or the CEBPA axis or both. Thus, vitamin A might be used to treat obesity and its related diseases.
Collapse
Affiliation(s)
- Linjun Xie
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Nutrition and Health, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- National Clinical Research Center for Child Health and Disorder, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Liying Zou
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Nutrition and Health, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- National Clinical Research Center for Child Health and Disorder, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Jie Chen
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Nutrition and Health, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- National Clinical Research Center for Child Health and Disorder, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Youxue Liu
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Nutrition and Health, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- National Clinical Research Center for Child Health and Disorder, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| |
Collapse
|
60
|
Jo YH, Peng DQ, Kim WS, Kim SJ, Kim NY, Kim SH, Ghassemi Nejad J, Lee JS, Lee HG. The effects of vitamin A supplementation during late-stage pregnancy on longissimus dorsi muscle tissue development, birth traits, and growth performance in postnatal Korean native calves. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2019; 33:742-752. [PMID: 32054186 PMCID: PMC7206403 DOI: 10.5713/ajas.19.0413] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 11/18/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE This study investigated the effects of vitamin A (VA) supplementation during late-stage pregnancy on longissimus dorsi muscle tissue development, birth traits, and growth performance of postnatal Korean native calves. METHODS In the preliminary experiment, twenty-six pregnant cattle (initial body weight [BW] = 319 kg (standard deviation [SD] = 30.1; 1st parity) were randomly assigned to the control and treatment groups. The treatment group received VA supplementation at 24,000 IU/d from gestational day 225 until delivery. In the main experiment, twelve pregnant cattle (initial BW = 317 kg [SD = 31.3]; 1st parity) were treated with VA supplementation at 24,000 IU/d (gestational days 150 to 225) and at 78,000 IU/d (gestational day 225 until delivery). Serum VA levels were analyzed in pregnant cattle, and the growth performance, gene expression, and serum VA levels were analyzed in the offspring. RESULTS Serum VA levels in pregnant cattle decreased the late gestation in both experiments (p<0.001). In the main experiment, pregnant cattle at parturition and offspring at birth in the treatment group had higher serum VA levels than those in the control group (p<0.05). In the treatment groups, an increased birth weight was observed in the main experimental group (p = 0.022), and a tendency (p = 0.088) toward an increased birth weight was observed in the preliminary experimental group. However, no differences were observed in the feed intake, average daily gain, gain-to-feed ratio, or BW of 31-day-old calves. Gene expression was analyzed in longissimus dorsi muscles of 31-day-old calves. VA supplementation in pregnant cattle stimulated postnatal muscle development in offspring by elevating myogenic factor 5 (MYF5), MYF6, and myoblast determination levels (p<0.05). Moreover, preadipocyte-related marker genes such as extracellular signal-regulated kinase 2 and krüppel-like factor 2 were higher in the treatment group than in the control group (p<0.05). CONCLUSION VA supplementation (78,000 IU/d) in late-stage pregnant cattle maintained serum VA levels. In addition, 78,000 IU/d VA supplementation increased the birth weight and expression of genes related to muscle and preadipocyte development in offspring. Overall, 78,000 IU/d VA supplementation in pregnant cattle is beneficial to newborn calves.
Collapse
Affiliation(s)
- Yong Ho Jo
- Department of Animal Science and Technology, Konkuk University, Seoul 05029, Korea.,Team of an Educational Program for Specialists in Global Animal Science, Brain Korea 21 Plus, Konkuk University, Seoul 05029, Korea
| | - Dong Qiao Peng
- Department of Animal Science and Technology, Konkuk University, Seoul 05029, Korea.,Team of an Educational Program for Specialists in Global Animal Science, Brain Korea 21 Plus, Konkuk University, Seoul 05029, Korea
| | - Won Seob Kim
- Department of Animal Science and Technology, Konkuk University, Seoul 05029, Korea.,Team of an Educational Program for Specialists in Global Animal Science, Brain Korea 21 Plus, Konkuk University, Seoul 05029, Korea
| | - Seong Jin Kim
- Asia Pacific Ruminant Institute, Icheon 467814, Korea
| | - Na Yeon Kim
- Asia Pacific Ruminant Institute, Icheon 467814, Korea
| | - Sung Hak Kim
- Department of Animal Science, Chonnam National University, Gwangju 61186, Korea
| | - Jalil Ghassemi Nejad
- Department of Animal Science and Technology, Konkuk University, Seoul 05029, Korea.,Team of an Educational Program for Specialists in Global Animal Science, Brain Korea 21 Plus, Konkuk University, Seoul 05029, Korea
| | - Jae Sung Lee
- Department of Animal Science and Technology, Konkuk University, Seoul 05029, Korea
| | - Hong Gu Lee
- Department of Animal Science and Technology, Konkuk University, Seoul 05029, Korea.,Team of an Educational Program for Specialists in Global Animal Science, Brain Korea 21 Plus, Konkuk University, Seoul 05029, Korea
| |
Collapse
|
61
|
Busnelli M, Manzini S, Bonacina F, Soldati S, Barbieri SS, Amadio P, Sandrini L, Arnaboldi F, Donetti E, Laaksonen R, Paltrinieri S, Scanziani E, Chiesa G. Fenretinide treatment accelerates atherosclerosis development in apoE-deficient mice in spite of beneficial metabolic effects. Br J Pharmacol 2019; 177:328-345. [PMID: 31621898 DOI: 10.1111/bph.14869] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 09/03/2019] [Accepted: 09/07/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Fenretinide, a synthetic retinoid derivative first investigated for cancer prevention and treatment, has been shown to ameliorate glucose tolerance, improve plasma lipid profile and reduce body fat mass. These effects, together with its ability to inhibit ceramide synthesis, suggest that fenretinide may have an anti-atherosclerotic action. EXPERIMENTAL APPROACH To this aim, nine-week-old apoE-knockout (EKO) female mice were fed for twelve weeks a Western diet, without (control) or with (0.1% w/w) fenretinide. As a reference, wild-type (WT) mice were treated similarly. Growth and metabolic parameters were monitored throughout the study. Atherosclerosis development was evaluated in the aorta and at the aortic sinus. Blood and lymphoid organs were further characterized with thorough cytological/histological and immunocytofluorimetric analyses. KEY RESULTS Fenretinide treatment significantly lowered body weight, glucose levels and plasma levels of total cholesterol, triglycerides, and phospholipids. In the liver, fenretinide remarkably reduced hepatic glycogenosis and steatosis driven by the Western diet. Treated spleens were abnormally enlarged, with severe follicular atrophy and massive extramedullary haematopoiesis. Severe renal hemosiderin deposition was observed in treated EKO mice. Treatment resulted in a threefold increase of total leukocytes (WT and EKO) and raised the activated/resting monocyte ratio in EKO mice. Finally, atherosclerosis development was markedly increased at the aortic arch, thoracic and abdominal aorta of fenretinide-treated mice. CONCLUSIONS AND IMPLICATIONS We provide the first evidence that, despite beneficial metabolic effects, fenretinide treatment may enhance the development of atherosclerosis.
Collapse
Affiliation(s)
- Marco Busnelli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Stefano Manzini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Fabrizia Bonacina
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Sabina Soldati
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy
| | | | | | - Leonardo Sandrini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.,IRCCS, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Francesca Arnaboldi
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, Italy
| | - Elena Donetti
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, Italy
| | - Reijo Laaksonen
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Saverio Paltrinieri
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy
| | - Eugenio Scanziani
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy.,Mouse and Animal Pathology Laboratory (MAPLab), Fondazione UniMi, Milan, Italy
| | - Giulia Chiesa
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
62
|
Abedi-Taleb E, Vahabi Z, Sekhavati-Moghadam E, Khedmat L, Jazayeri S, Saboor-Yaraghi AA. Upregulation of FNDC5 gene expression in C2C12 cells after single and combined treatments of resveratrol and ATRA. Lipids Health Dis 2019; 18:181. [PMID: 31640715 PMCID: PMC6806552 DOI: 10.1186/s12944-019-1128-y] [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: 05/20/2019] [Accepted: 10/03/2019] [Indexed: 01/18/2023] Open
Abstract
Background Irisin is a newly discovered myokine that secreted from skeletal muscle cells. Several studies showed that irisin involves in thermogenesis and increases the expression of browning markers such as uncoupling protein-1 that in turns induces the conversion of white adipose tissue to brown fat. Resveratrol (Res) and all-trans retinoic acid (ATRA) can also upregulate the expression of thermogenesis genes. In the present study, the effects of single and combined treatments of Res and ATRA on fibronectin type III domain containing 5 (FNDC5) gene expression was explored. Methods The mouse myoblasts, C2C12 cells, were seeded in 6-well plastic plates and cultured in DMEM media. After differentiation, in a pilot study, C2C12 myotubes were treated with different concentrations of Res and ATRA for 12 h. The best result was obtained by treatment of 1and 25 μM of Res and 1 μM of ATRA. Then the main study was continued by single and combined treatment of these compounds at chosen concentration. After treatments, total RNA was extracted from C2C12 cells. Complementary DNA (cDNA) was generated by the cDNA synthesis kit and FNDC5 mRNA expression was evaluated by the real-time PCR method. Results The FNDC5 gene expression in C2C12 myotubes of alone-treated with 1 μM, 25 μM Res and 10 μM ATRA did not change compared to vehicle group. However, in combination-treated the expression of FNDC5 gene was significantly increased compared to vehicle group. Conclusion This is the first evidence that Res and ATRA can regulate FNDC5 gene expression in C2C12 myotubes. More investigations are necessary to explore the therapeutic effects of these nutrients in obesity, diabetes, cardiac and neurovascular disease.
Collapse
Affiliation(s)
- Elahe Abedi-Taleb
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Vahabi
- Department of Geriatric Medicine, Ziaeian Hospital, Tehran University of Medical Sciences, Tehran, Iran.,Memory and Behavioral Neurology Division, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Leila Khedmat
- Health Management Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Shima Jazayeri
- Department of Nutrition and Biochemistry, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Akbar Saboor-Yaraghi
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran. .,Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, 141613151, Iran.
| |
Collapse
|
63
|
Liggins MC, Li F, Zhang LJ, Dokoshi T, Gallo RL. Retinoids Enhance the Expression of Cathelicidin Antimicrobial Peptide during Reactive Dermal Adipogenesis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 203:1589-1597. [PMID: 31420464 PMCID: PMC9233297 DOI: 10.4049/jimmunol.1900520] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 07/16/2019] [Indexed: 11/30/2023]
Abstract
A subset of dermal fibroblasts undergo rapid differentiation into adipocytes in response to infection and acutely produce the cathelicidin antimicrobial peptide gene Camp Vitamin A and other retinoids inhibit adipogenesis yet can show benefit to skin disorders, such as cystic acne, that are exacerbated by bacteria. We observed that retinoids potently increase and sustain the expression of Camp in preadipocytes undergoing adipogenesis despite inhibition of markers of adipogenesis, such as Adipoq, Fabp4, and Rstn Retinoids increase cathelicidin in both mouse and human preadipocytes, but this enhancement of antimicrobial peptide expression did not occur in keratinocytes or a sebocyte cell line. Preadipocytes undergoing adipogenesis more effectively inhibited growth of Staphylococcus aureus when exposed to retinoic acid. Whole transcriptome analysis identified hypoxia-inducible factor 1-α (HIF-1α) as a mechanism through which retinoids mediate this response. These observations uncouple the lipid accumulation element of adipogenesis from the innate immune response and uncover a mechanism, to our knowledge previously unsuspected, that may explain therapeutic benefits of retinoids in some skin disorders.
Collapse
Affiliation(s)
- Marc C Liggins
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093; and
| | - Fengwu Li
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093; and
| | - Ling-Juan Zhang
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093; and
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Tatsuya Dokoshi
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093; and
| | - Richard L Gallo
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093; and
| |
Collapse
|
64
|
Lee SA, Jiang H, Feranil JB, Brun PJ, Blaner WS. Adipocyte-specific expression of a retinoic acid receptor α dominant negative form causes glucose intolerance and hepatic steatosis in mice. Biochem Biophys Res Commun 2019; 514:1231-1237. [PMID: 31109648 DOI: 10.1016/j.bbrc.2019.05.075] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 05/09/2019] [Indexed: 11/30/2022]
Abstract
All-trans-retinoic acid (ATRA) has been well described as a positive regulator for early stage of adipocyte differentiation and lipid metabolism and also linked to an in vivo fat-lowering effect in mice. However, not all studies support this association. Our objective was to characterize the action of ATRA in mature adipocytes of mice by ablating RAR signaling through overexpression of a well-characterized dominant negative RARα mutant (RARdn) form specifically in adipocytes. Altered RAR signaling in adipocytes resulted in a significant decrease in ATRA levels in visceral and brown adipose tissues as well as liver tissue. This was linked to significant impairments in glucose clearance and elevated hepatic lipid accumulation for chow diet fed mice, indicating the development of metabolic disease, including hepatic steatosis. In addition, we found that adipose RARdn expression in mice fed a chow diet decreased thermogenesis. We conclude that altered RAR signaling and ATRA levels in adipocytes impacts glucose and lipid metabolism in mice.
Collapse
Affiliation(s)
- Seung-Ah Lee
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, United States; Present Address: Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea.
| | - Hongfeng Jiang
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, United States.
| | - Jun B Feranil
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, United States.
| | - Pierre-Jacques Brun
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, United States.
| | - William S Blaner
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, United States.
| |
Collapse
|
65
|
Suzuki M, Chen HJ, Tomonaga S, Hashimoto O, Kawada T, Matsui T, Funaba M. Chronic retinoic acid treatment induces differentiation and changes in the metabolite levels of brown (pre)adipocytes. Cell Biochem Funct 2019; 37:377-384. [PMID: 31215681 DOI: 10.1002/cbf.3416] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 04/11/2019] [Accepted: 05/08/2019] [Indexed: 12/19/2022]
Abstract
Dietary vitamin A status affects energy metabolism. The present study explored the effect of all-trans retinoic acid (ATRA) on the expression levels of molecules and metabolites of brown adipocytes. Chronic ATRA treatment was initiated during the early stage (days 0-8) or late stage (days 8-12) of adipogenesis. Treatment with ATRA during the early and late stage of adipogenesis resulted in an increase in the expression level of Ucp1 and Cidea, genes highly expressed in brown adipocytes, on day 8 and day 12, respectively, whereas expression of Pgc-1α, another gene expressed during brown adipogenesis, was unaffected by ATRA. Non-targeted metabolomic analyses indicated that the pathways related to the glucose metabolism were affected by ATRA, irrespective of the differentiation stage. Cellular levels of glucose 6-phosphate, fructose 6-phosphate, citric acid, and succinic acid decreased after ATRA treatment on days 8 and 12. In contrast, glucose level was higher in ATRA-treated cells on day 8, but it was lower on day 12. ATRA decreased the cellular level of aconitic acid, fumaric acid, and malic acid on day 12 but not on day 8. Furthermore, ATRA increased the expression level of Hxk2 and downregulated the expressions of G6pdh and Pfkl/Pfkp on day 8 but not on day 12. Together, the results indicate that the chronic treatment with ATRA stimulated the formation of activated brown adipocytes, eventually leading to alterations in the levels of cellular metabolites related to glucose metabolism. SIGNIFICANCE OF THE STUDY: Significance of the study treatment with all-trans retinoic acid (ATRA) during the early and late stage of adipogenesis increased the expression of Ucp1 and Cidea, genes highly expressed in brown adipocytes, on day 8 and day 12. Cellular levels of glucose 6-phosphate, fructose 6-phosphate, citric acid, and succinic acid decreased after ATRA treatment on days 8 and 12. In contrast, glucose level was higher in ATRA-treated cells on day 8, but it was lower on day 12. The present results indicate that ATRA stimulated the formation of activated brown adipocytes, eventually leading to alterations in the levels of cellular metabolites related to glucose metabolism.
Collapse
Affiliation(s)
- Mika Suzuki
- Division of Applied Biosciences, Kyoto University Graduate School of Agriculture, Kyoto, Japan
| | - Hsuan-Ju Chen
- Division of Applied Biosciences, Kyoto University Graduate School of Agriculture, Kyoto, Japan
| | - Shozo Tomonaga
- Division of Applied Biosciences, Kyoto University Graduate School of Agriculture, Kyoto, Japan
| | - Osamu Hashimoto
- Laboratory of Experimental Animal Science, Kitasato University School of Veterinary Medicine, Towada, Japan
| | - Teruo Kawada
- Division of Food Science and Biotechnology, Kyoto University Graduate School of Agriculture, Kyoto, Japan
| | - Tohru Matsui
- Division of Applied Biosciences, Kyoto University Graduate School of Agriculture, Kyoto, Japan
| | - Masayuki Funaba
- Division of Applied Biosciences, Kyoto University Graduate School of Agriculture, Kyoto, Japan
| |
Collapse
|
66
|
Cellular retinoic acid binding protein 1 protects mice from high-fat diet-induced obesity by decreasing adipocyte hypertrophy. Int J Obes (Lond) 2019; 44:466-474. [PMID: 31164723 PMCID: PMC6891142 DOI: 10.1038/s41366-019-0379-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 03/07/2019] [Accepted: 04/01/2019] [Indexed: 12/21/2022]
Abstract
Objectives Obesity, an emerging global health issue, involves numerous factors; understanding its underlying mechanisms for prevention and therapeutics is urgently needed. Cellular retinoic acid binding protein 1 (Crabp1) knockout (CKO) mice exhibit an obese phenotype under normal diet feedings, which prompted us to propose that Crabp1 could play a role in modulating adipose tissue development/homeostasis. Studies were designed to elucidate the underlying mechanism of Crabp1’s action in reducing obesity. Subjects/methods In animal studies, 6 weeks old male wild type and CKO mice were fed with normal diet (ND) or high fat diet (HFD) for 10 weeks. Body weight and food intake were regularly monitored. Glucose tolerance test and biological parameters of plasma (glucose and insulin levels) were measured after 10 weeks of ND vs. HFD feedings. Visceral adipose tissues were collected for histological and molecular analyses to determine affected signaling pathways. In cell culture studies, the 3T3L1 adipocyte differentiation model was used to examine and validate relevant signaling pathways. Results CKO mice, compared to WT mice, gained more body weight, exhibited more elevated fasting plasma glucose levels, and developed more severe impaired glucose tolerance under both ND and HFD. Histological examination revealed readily increased adipocyte hypertrophy and adipose tissue inflammation under HFD feedings. In 3T3L1 adipocytes, Crabp1 silencing enhanced extracellular signal-regulated kinase 1/2 (ERK1/2) activation, accompanied by elevated markers and signaling pathways of lipid accumulation and adipocyte hypertrophy. Conclusions This study identifies Crabp1’s physiological role against the development of obesity. The protective function of CRABP1 is likely attributed to its classically proposed (canonical) activity as a trap for RA, which will reduce RA availability, thereby dampening RA-stimulated ERK1/2 activation and adipocyte hypertrophy. The results suggest Crabp1 as a potentially new therapeutic target in managing obesity and metabolic diseases.
Collapse
|
67
|
Krois CR, Vuckovic MG, Huang P, Zaversnik C, Liu CS, Gibson CE, Wheeler MR, Obrochta KM, Min JH, Herber CB, Thompson AC, Shah ID, Gordon SP, Hellerstein MK, Napoli JL. RDH1 suppresses adiposity by promoting brown adipose adaptation to fasting and re-feeding. Cell Mol Life Sci 2019; 76:2425-2447. [PMID: 30788515 PMCID: PMC6531335 DOI: 10.1007/s00018-019-03046-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 02/07/2019] [Accepted: 02/11/2019] [Indexed: 12/26/2022]
Abstract
RDH1 is one of the several enzymes that catalyze the first of the two reactions to convert retinol into all-trans-retinoic acid (atRA). Here, we show that Rdh1-null mice fed a low-fat diet gain more weight as adiposity (17% males, 13% females) than wild-type mice by 20 weeks old, despite neither consuming more calories nor decreasing activity. Glucose intolerance and insulin resistance develop following increased adiposity. Despite the increase in white fat pads, epididymal white adipose does not express Rdh1, nor does muscle. Brown adipose tissue (BAT) and liver express Rdh1 at relatively high levels compared to other tissues. Rdh1 ablation lowered body temperatures during ambient conditions. Given the decreased body temperature, we focused on BAT. A lack of differences in BAT adipogenic gene expression between Rdh1-null mice and wild-type mice, including Pparg, Prdm16, Zfp516 and Zfp521, indicated that the phenotype was not driven by brown adipose hyperplasia. Rather, Rdh1 ablation eliminated the increase in BAT atRA that occurs after re-feeding. This disruption of atRA homeostasis increased fatty acid uptake, but attenuated lipolysis in primary brown adipocytes, resulting in increased lipid content and larger lipid droplets. Rdh1 ablation also decreased mitochondrial proteins, including CYCS and UCP1, the mitochondria oxygen consumption rate, and disrupted the mitochondria membrane potential, further reflecting impaired BAT function, resulting in both BAT and white adipose hypertrophy. RNAseq revealed dysregulation of 424 BAT genes in null mice, which segregated predominantly into differences after fasting vs after re-feeding. Exceptions were Rbp4 and Gbp2b, which increased during both dietary conditions. Rbp4 encodes the serum retinol-binding protein-an insulin desensitizer. Gbp2b encodes a GTPase. Because Gbp2b increased several hundred-fold, we overexpressed it in brown adipocytes. This caused a shift to larger lipid droplets, suggesting that GBP2b affects signaling downstream of the β-adrenergic receptor during basal thermogenesis. Thus, Rdh1-generated atRA in BAT regulates multiple genes that promote BAT adaptation to whole-body energy status, such as fasting and re-feeding. These gene expression changes promote optimum mitochondria function and thermogenesis, limiting adiposity. Attenuation of adiposity and insulin resistance suggests that RDH1 mitigates metabolic syndrome.
Collapse
Affiliation(s)
- Charles R Krois
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, 119 Morgan Hall, Berkeley, CA, 94720-3104, USA
- Department of Chemistry and Geology, Minnesota State University, 241 Ford Hall, Mankato, MN, 56001, USA
| | - Marta G Vuckovic
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, 119 Morgan Hall, Berkeley, CA, 94720-3104, USA
| | - Priscilla Huang
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, 119 Morgan Hall, Berkeley, CA, 94720-3104, USA
- Arizona College of Osteopathic Medicine, Midwestern University, 19555 North 59th Avenue, Glendale, AZ, 85308, USA
| | - Claire Zaversnik
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, 119 Morgan Hall, Berkeley, CA, 94720-3104, USA
- AgroSup Dijon, 26 Bd Petitjean, 21000, Dijon, France
| | - Conan S Liu
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, 119 Morgan Hall, Berkeley, CA, 94720-3104, USA
- Sidney Kimmel Medical College, 1025 Walnut Street, Philadelphia, PA, 19104, USA
| | - Candice E Gibson
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, 119 Morgan Hall, Berkeley, CA, 94720-3104, USA
| | - Madelyn R Wheeler
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, 119 Morgan Hall, Berkeley, CA, 94720-3104, USA
- UC Davis School of Medicine, 4102 Sherman Way, Sacramento, CA, 95817, USA
| | - Kristin M Obrochta
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, 119 Morgan Hall, Berkeley, CA, 94720-3104, USA
- Biomarin Pharmaceutical Inc., 105 Digital Drive, Novato, CA, 94949, USA
| | - Jin H Min
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, 119 Morgan Hall, Berkeley, CA, 94720-3104, USA
- Nova Southeastern University, 3301 College Avenue, Fort Lauderdale, FL, 33314, USA
| | - Candice B Herber
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, 119 Morgan Hall, Berkeley, CA, 94720-3104, USA
- University of California, San Francisco, Rock Hall 281, 1550 4th Street, San Francisco, CA, 94158, USA
| | - Airlia C Thompson
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, 119 Morgan Hall, Berkeley, CA, 94720-3104, USA
- Stanford University, Lorry Lokey Building Room 164, 337 Campus Drive, Stanford, CA, 94305-5020, USA
| | - Ishan D Shah
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, 119 Morgan Hall, Berkeley, CA, 94720-3104, USA
- Keck School of Medicine, University of Southern California, 1975 Zonal Avenue, Keith Administration (KAM) 100, Los Angeles, CA, 90089-9020, USA
| | - Sean P Gordon
- DOE Joint Genome Institute, 2800 Mitchell Dr # 100, Walnut Creek, CA, 94598, USA
| | - Marc K Hellerstein
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, 119 Morgan Hall, Berkeley, CA, 94720-3104, USA
| | - Joseph L Napoli
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, 119 Morgan Hall, Berkeley, CA, 94720-3104, USA.
| |
Collapse
|
68
|
Marinkovic M, Fuoco C, Sacco F, Cerquone Perpetuini A, Giuliani G, Micarelli E, Pavlidou T, Petrilli LL, Reggio A, Riccio F, Spada F, Vumbaca S, Zuccotti A, Castagnoli L, Mann M, Gargioli C, Cesareni G. Fibro-adipogenic progenitors of dystrophic mice are insensitive to NOTCH regulation of adipogenesis. Life Sci Alliance 2019; 2:e201900437. [PMID: 31239312 PMCID: PMC6599969 DOI: 10.26508/lsa.201900437] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 06/07/2019] [Indexed: 12/14/2022] Open
Abstract
Fibro-adipogenic progenitors (FAPs) promote satellite cell differentiation in adult skeletal muscle regeneration. However, in pathological conditions, FAPs are responsible for fibrosis and fatty infiltrations. Here we show that the NOTCH pathway negatively modulates FAP differentiation both in vitro and in vivo. However, FAPs isolated from young dystrophin-deficient mdx mice are insensitive to this control mechanism. An unbiased mass spectrometry-based proteomic analysis of FAPs from muscles of wild-type and mdx mice suggested that the synergistic cooperation between NOTCH and inflammatory signals controls FAP differentiation. Remarkably, we demonstrated that factors released by hematopoietic cells restore the sensitivity to NOTCH adipogenic inhibition in mdx FAPs. These results offer a basis for rationalizing pathological ectopic fat infiltrations in skeletal muscle and may suggest new therapeutic strategies to mitigate the detrimental effects of fat depositions in muscles of dystrophic patients.
Collapse
Affiliation(s)
| | - Claudia Fuoco
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Francesca Sacco
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | | | - Giulio Giuliani
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Elisa Micarelli
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | | | | | - Alessio Reggio
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Federica Riccio
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Filomena Spada
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Simone Vumbaca
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | | | - Luisa Castagnoli
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Cesare Gargioli
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Gianni Cesareni
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
69
|
Abstract
Much evidence has accumulated in the literature over the last fifteen years that indicates vitamin A has a role in metabolic disease prevention and causation. This literature proposes that vitamin A can affect obesity development and the development of obesity-related diseases including insulin resistance, type 2 diabetes, hepatic steatosis and steatohepatitis, and cardiovascular disease. Retinoic acid, the transcriptionally active form of vitamin A, accounts for many of the reported associations. However, a number of proteins involved in vitamin A metabolism, including retinol-binding protein 4 (RBP4) and aldehyde dehydrogenase 1A1 (ALDH1A1, alternatively known as retinaldehyde dehydrogenase 1 or RALDH1), have also been identified as being associated with metabolic disease. Some of the reported effects of these vitamin A-related proteins are proposed to be independent of their roles in assuring normal retinoic acid homeostasis. This review will consider both human observational data as well as published data from molecular studies undertaken in rodent models and in cells in culture. The primary focus of the review will be on the effects that vitamin A per se and proteins involved in vitamin A metabolism have on adipocytes, adipose tissue biology, and adipose-related disease, as well as on early stage liver disease, including non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH).
Collapse
Affiliation(s)
- William S Blaner
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York 10032.
| |
Collapse
|
70
|
Tezze C, Romanello V, Sandri M. FGF21 as Modulator of Metabolism in Health and Disease. Front Physiol 2019; 10:419. [PMID: 31057418 PMCID: PMC6478891 DOI: 10.3389/fphys.2019.00419] [Citation(s) in RCA: 216] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 03/27/2019] [Indexed: 12/12/2022] Open
Abstract
Fibroblast growth factor 21 (FGF21) is a hormone that regulates important metabolic pathways. FGF21 is expressed in several metabolically active organs and interacts with different tissues. The FGF21 function is complicated and well debated due to its different sites of production and actions. Striated muscles are plastic tissues that undergo adaptive changes within their structural and functional properties in order to meet their different stresses, recently, they have been found to be an important source of FGF21. The FGF21 expression and secretion from skeletal muscles happen in both mouse and in humans during their different physiological and pathological conditions, including exercise and mitochondrial dysfunction. In this review, we will discuss the recent findings that identify FG21 as beneficial and/or detrimental cytokine interacting as an autocrine or endocrine in order to modulate cellular function, metabolism, and senescence.
Collapse
Affiliation(s)
- Caterina Tezze
- Veneto Institute of Molecular Medicine, Padua, Italy.,Department of Biomedical Science, University of Padua, Padua, Italy
| | - Vanina Romanello
- Veneto Institute of Molecular Medicine, Padua, Italy.,Department of Biomedical Science, University of Padua, Padua, Italy
| | - Marco Sandri
- Veneto Institute of Molecular Medicine, Padua, Italy.,Department of Biomedical Science, University of Padua, Padua, Italy.,Department of Medicine, McGill University, Montreal, QC, Canada.,Department of Biomedical Science, Myology Center, University of Padua, Padua, Italy
| |
Collapse
|
71
|
Coronel J, Pinos I, Amengual J. β-carotene in Obesity Research: Technical Considerations and Current Status of the Field. Nutrients 2019; 11:E842. [PMID: 31013923 PMCID: PMC6521044 DOI: 10.3390/nu11040842] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 04/02/2019] [Accepted: 04/06/2019] [Indexed: 12/21/2022] Open
Abstract
Over the past decades, obesity has become a rising health problem as the accessibility to high calorie, low nutritional value food has increased. Research shows that some bioactive components in fruits and vegetables, such as carotenoids, could contribute to the prevention and treatment of obesity. Some of these carotenoids are responsible for vitamin A production, a hormone-like vitamin with pleiotropic effects in mammals. Among these effects, vitamin A is a potent regulator of adipose tissue development, and is therefore important for obesity. This review focuses on the role of the provitamin A carotenoid β-carotene in human health, emphasizing the mechanisms by which this compound and its derivatives regulate adipocyte biology. It also discusses the physiological relevance of carotenoid accumulation, the implication of the carotenoid-cleaving enzymes, and the technical difficulties and considerations researchers must take when working with these bioactive molecules. Thanks to the broad spectrum of functions carotenoids have in modern nutrition and health, it is necessary to understand their benefits regarding to metabolic diseases such as obesity in order to evaluate their applicability to the medical and pharmaceutical fields.
Collapse
Affiliation(s)
- Johana Coronel
- Department of Food Sciences and Human Nutrition, University of Illinois Urbana Champaign, Urbana, IL 61801, USA.
| | - Ivan Pinos
- Division of Nutritional Sciences, University of Illinois Urbana Champaign, Urbana, IL 61801, USA.
| | - Jaume Amengual
- Department of Food Sciences and Human Nutrition, University of Illinois Urbana Champaign, Urbana, IL 61801, USA.
- Division of Nutritional Sciences, University of Illinois Urbana Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
72
|
Small molecules for fat combustion: targeting obesity. Acta Pharm Sin B 2019; 9:220-236. [PMID: 30976490 PMCID: PMC6438825 DOI: 10.1016/j.apsb.2018.09.007] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/01/2018] [Accepted: 08/22/2018] [Indexed: 12/11/2022] Open
Abstract
Obesity is increasing in an alarming rate worldwide, which causes higher risks of some diseases, such as type 2 diabetes, cardiovascular diseases, and cancer. Current therapeutic approaches, either pancreatic lipase inhibitors or appetite suppressors, are generally of limited effectiveness. Brown adipose tissue (BAT) and beige cells dissipate fatty acids as heat to maintain body temperature, termed non-shivering thermogenesis; the activity and mass of BAT and beige cells are negatively correlated with overweight and obesity. The existence of BAT and beige cells in human adults provides an effective weight reduction therapy, a process likely to be amenable to pharmacological intervention. Herein, we combed through the physiology of thermogenesis and the role of BAT and beige cells in combating with obesity. We summarized the thermogenic regulators identified in the past decades, targeting G protein-coupled receptors, transient receptor potential channels, nuclear receptors and miscellaneous pathways. Advances in clinical trials were also presented. The main purpose of this review is to provide a comprehensive and up-to-date knowledge from the biological importance of thermogenesis in energy homeostasis to the representative thermogenic regulators for treating obesity. Thermogenic regulators might have a large potential for further investigations to be developed as lead compounds in fighting obesity.
Collapse
Key Words
- AKT, protein kinase B
- ALDH9, aldehyde dehydrogenase 9
- AMPK, AMP-activated protein kinase
- ATP, adenosine triphosphate
- BA, bile acids
- BAT, brown adipose tissue
- BMP8b, bone morphogenetic protein 8b
- Beige cells
- Brown adipose tissue
- C/EBPα, CCAAT/enhancer binding protein α
- CLA, cis-12 conjugated linoleic acid
- CRABP-II, cellular RA binding protein type II
- CRE, cAMP response element
- Cidea, cell death-inducing DNA fragmentation factor α-like effector A
- Dio2, iodothyronine deiodinase type 2
- ERE, estrogen response element
- ERs, estrogen receptors
- FAS, fatty acid synthase
- FGF21, fibroblast growth factor 21
- GPCRs, G protein-coupled receptors
- HFD, high fat diet
- LXR, liver X receptors
- MAPK, mitogen-activated protein kinase
- OXPHOS, oxidative phosphorylation
- Obesity
- PDEs, phosphodiesterases
- PET-CT, positron emission tomography combined with computed tomography
- PGC-1α, peroxisome proliferator-activated receptor γ coactivator 1-α
- PKA, protein kinase A
- PPARs, peroxisome proliferator-activated receptors
- PPREs, peroxisome proliferator response elements
- PRDM16, PR domain containing 16
- PTP1B, protein-tyrosine phosphatase 1B
- PXR, pregnane X receptor
- RA, retinoic acid
- RAR, RA receptor
- RARE, RA response element
- RMR, resting metabolic rate
- RXR, retinoid X receptor
- SIRT1, silent mating type information regulation 2 homolog 1
- SNS, sympathetic nervous system
- TFAM, mitochondrial transcription factor A
- TMEM26, transmembrane protein 26
- TRPs, transient receptor potential cation channels
- Thermogenesis
- UCP1, uncoupling protein 1
- Uncoupling protein 1
- VDR, vitamin D receptor
- VDRE, VDR response elements
- WAT, white adipose tissue
- cAMP, cyclic adenosine monophosphate
- cGMP, cyclic guanosine monophosphate
- β3-AR, β3-adrenergic receptor
Collapse
|
73
|
Choe U, Yu LL, Wang TTY. The Science behind Microgreens as an Exciting New Food for the 21st Century. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:11519-11530. [PMID: 30343573 DOI: 10.1021/acs.jafc.8b03096] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Chronic diseases are a major health problem in the United States. Accumulated data suggest that consumption of vegetables can significantly reduce the risk of many chronic diseases. Dietary guidelines for 2015-2020 from the U.S. Department of Agriculture and the U.S. Department of Health and Human Services recommend 1-4 cups of vegetables per day for males and 1-3 cups of vegetables per day for females, depending on their age. However, the average intake of vegetables is below the recommended levels. Microgreens are young vegetable greens. Although they are small, microgreens have delicate textures, distinctive flavors, and various nutrients. In general, microgreens contain greater amounts of nutrients and health-promoting micronutrients than their mature counterparts. Because microgreens are rich in nutrients, smaller amounts may provide similar nutritional effects compared to larger quantities of mature vegetables. However, literature on microgreens remains limited. In this Review, we discuss chemical compositions, growing conditions, and biological efficacies of microgreens. We seek to stimulate interest in further study of microgreens as a promising dietary component for potential use in diet-based disease prevention.
Collapse
Affiliation(s)
- Uyory Choe
- Department of Nutrition and Food Science , University of Maryland , College Park , Maryland 20742 , United States
- Diet, Genomics and Immunology Laboratory, Beltsville Human Nutrition Research Center, ARS , U.S. Department of Agriculture , 10300 Baltimore Avenue , Beltsville , Maryland 20705 , United States
| | - Liangli Lucy Yu
- Department of Nutrition and Food Science , University of Maryland , College Park , Maryland 20742 , United States
| | - Thomas T Y Wang
- Diet, Genomics and Immunology Laboratory, Beltsville Human Nutrition Research Center, ARS , U.S. Department of Agriculture , 10300 Baltimore Avenue , Beltsville , Maryland 20705 , United States
| |
Collapse
|
74
|
Wang B, Fu X, Zhu MJ, Du M. Retinoic acid inhibits white adipogenesis by disrupting GADD45A-mediated Zfp423 DNA demethylation. J Mol Cell Biol 2018; 9:338-349. [PMID: 28992291 DOI: 10.1093/jmcb/mjx026] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 08/01/2017] [Indexed: 12/20/2022] Open
Abstract
Retinoic acid (RA), a bioactive metabolite of vitamin A, is a critical mediator of cell differentiation. RA blocks adipogenesis, but mechanisms remain to be established. ZFP423 is a key transcription factor maintaining white adipose identity. We found that RA inhibits Zfp423 expression and adipogenesis via blocking DNA demethylation in the promoter of Zfp423, a process mediated by growth arrest and DNA-damage-inducible protein alpha (GADD45A). RA induces the partnering between retinoic acid receptor (RAR) and tumor suppressor inhibitor of growth protein 1 (ING1), which prevents the formation of GADD45A and ING1 complex necessary for locus-specific Zfp423 DNA demethylation. In vivo, vitamin A supplementation prevents obesity, downregulates Gadd45a expression, and reduces GADD45A binding and DNA demethylation in the Zfp423 promoter. Inhibition of Zfp423 expression due to RA contributes to the enhanced brown adipogenesis. In summary, RA inhibits white adipogenesis by inducing RAR and ING1 interaction and inhibiting Gadd45a expression, which prevents GADD45A-mediated DNA demethylation.
Collapse
Affiliation(s)
- B Wang
- Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100094, China.,Department of Animal Sciences, Washington State University, Pullman, WA 99164, USA
| | - Xing Fu
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, USA
| | - Mei-Jun Zhu
- School of Food Science, Washington State University, Pullman, WA 99164, USA
| | - Min Du
- Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100094, China.,Department of Animal Sciences, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
75
|
Goudarzi F, Sarveazad A, Mahmoudi M, Mohammadalipour A, Chahardoli R, Malekshah OM, Karimi Gorgani S, Saboor-Yaraghi AA. Combined effect of retinoic acid and calcium on the in vitro differentiation of human adipose-derived stem cells to adipocytes. Arch Physiol Biochem 2018; 124:109-118. [PMID: 28850272 DOI: 10.1080/13813455.2017.1367009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
CONTEXT It has been shown that adipogenesis can be modulated by factors such as all-trans retinoic acid (ATRA) and calcium. OBJECTIVE To determine, the combined effect of ATRA and calcium on the differentiation of human adipose-derived stem cells (hADSCs). METHODS Mesenchymal stem cells (MSCs) were differentiated into the adipocytes by 0.5 and 1 µM of ATRA and 5 and 10 mM calcium separately or in combination. After MTS assay the differentiation of MSCs to adipocyte was evaluated, Oil Red O staining, GLUT4 concentration and gene expression of PPARG2, adiponectin, and GLUT4 were measured by Real-Time PCR. RESULTS Except 10 mM calcium treated group, other groups and more significantly combination treatments could reduce all adipocyte markers compared to the control. CONCLUSION These results suggest that ATRA and calcium together have significant inhibitory effect on adipogenesis that can be helpful for finding new mechanisms to prevent or control the adipogenesis.
Collapse
Affiliation(s)
- Farjam Goudarzi
- a Department of Biochemistry, School of Medicine , Hamadan University of Medical Sciences , Hamadan , Iran
| | - Arash Sarveazad
- b Colorectal Research Center , Iran University of Medical Sciences , Tehran , Iran
| | - Maryam Mahmoudi
- c Department of Cellular Molecular Nutrition, School of Nutritional Sciences and Dietetics , Tehran University of Medical Sciences , Tehran , Iran
| | - Adel Mohammadalipour
- a Department of Biochemistry, School of Medicine , Hamadan University of Medical Sciences , Hamadan , Iran
| | - Reza Chahardoli
- c Department of Cellular Molecular Nutrition, School of Nutritional Sciences and Dietetics , Tehran University of Medical Sciences , Tehran , Iran
| | - Obeid M Malekshah
- d Department of Pharmaceutics, Ernest Mario School of Pharmacy , Rutgers, The State University of New Jersey , Piscataway , NJ , USA
| | - Shiva Karimi Gorgani
- e Health Information Management Department , Paramedical School, Kermanshah University of Medical Sciences , Kermanshah , Iran
| | - Ali Akbar Saboor-Yaraghi
- f Department of Immunology, School of Public Health , Tehran University of Medical sciences , Tehran , Iran
| |
Collapse
|
76
|
Yang D, Vuckovic MG, Smullin CP, Kim M, Lo CPS, Devericks E, Yoo HS, Tintcheva M, Deng Y, Napoli JL. Modest Decreases in Endogenous All- trans-Retinoic Acid Produced by a Mouse Rdh10 Heterozygote Provoke Major Abnormalities in Adipogenesis and Lipid Metabolism. Diabetes 2018; 67:662-673. [PMID: 29321172 PMCID: PMC5860858 DOI: 10.2337/db17-0946] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 01/02/2018] [Indexed: 12/18/2022]
Abstract
Pharmacological dosing of all-trans-retinoic acid (atRA) controls adiposity in rodents by inhibiting adipogenesis and inducing fatty acid oxidation. Retinol dehydrogenases (Rdh) catalyze the first reaction that activates retinol into atRA. This study examined postnatal contributions of Rdh10 to atRA biosynthesis and physiological functions of endogenous atRA. Embryonic fibroblasts from Rdh10 heterozygote hypomorphs or with a total Rdh10 knockout exhibit decreased atRA biosynthesis and escalated adipogenesis. atRA or a retinoic acid receptor (RAR) pan-agonist reversed the phenotype. Eliminating one Rdh10 copy in vivo (Rdh10+/- ) yielded a modest decrease (≤25%) in the atRA concentration of liver and adipose but increased adiposity in male and female mice fed a high-fat diet (HFD); increased liver steatosis, glucose intolerance, and insulin resistance in males fed an HFD; and activated bone marrow adipocyte formation in females, regardless of dietary fat. Chronic dosing with low-dose atRA corrected the metabolic defects. These data resolve physiological actions of endogenous atRA, reveal sex-specific effects of atRA in vivo, and establish the importance of Rdh10 to metabolic control by atRA. The consequences of a modest decrease in tissue atRA suggest that impaired retinol activation may contribute to diabesity, and low-dose atRA therapy may ameliorate adiposity and its sequelae of glucose intolerance and insulin resistance.
Collapse
Affiliation(s)
- Di Yang
- Graduate Program in Metabolic Biology, University of California, Berkeley, Berkeley, CA
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA
| | - Marta G Vuckovic
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA
| | - Carolyn P Smullin
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA
| | - Myeongcheol Kim
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA
| | - Christabel Pui-See Lo
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA
| | - Emily Devericks
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA
| | - Hong Sik Yoo
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA
| | - Milena Tintcheva
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA
| | - Yinghua Deng
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA
| | - Joseph L Napoli
- Graduate Program in Metabolic Biology, University of California, Berkeley, Berkeley, CA
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA
| |
Collapse
|
77
|
Vitamin A and marbling attributes: Intramuscular fat hyperplasia effects in cattle. Meat Sci 2018; 137:139-146. [DOI: 10.1016/j.meatsci.2017.11.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 10/05/2017] [Accepted: 11/17/2017] [Indexed: 01/16/2023]
|
78
|
Abstract
Multiple binding and transport proteins facilitate many aspects of retinoid biology through effects on retinoid transport, cellular uptake, metabolism, and nuclear delivery. These include the serum retinol binding protein sRBP (aka Rbp4), the plasma membrane sRBP receptor Stra6, and the intracellular retinoid binding-proteins such as cellular retinol-binding proteins (CRBP) and cellular retinoic acid binding-proteins (CRABP). sRBP transports the highly lipophilic retinol through an aqueous medium. The major intracellular retinol-binding protein, CRBP1, likely enhances efficient retinoid use by providing a sink to facilitate retinol uptake from sRBP through the plasma membrane or via Stra6, delivering retinol or retinal to select enzymes that generate retinyl esters or retinoic acid, and protecting retinol/retinal from excess catabolism or opportunistic metabolism. Intracellular retinoic acid binding-proteins (CRABP1 and 2, and FABP5) seem to have more diverse functions distinctive to each, such as directing retinoic acid to catabolism, delivering retinoic acid to specific nuclear receptors, and generating non-canonical actions. Gene ablation of intracellular retinoid binding-proteins does not cause embryonic lethality or gross morphological defects. Metabolic and functional defects manifested in knockouts of CRBP1, CRBP2 and CRBP3, however, illustrate their essentiality to health, and in the case of CRBP2, to survival during limited dietary vitamin A. Future studies should continue to address the specific molecular interactions that occur between retinoid binding-proteins and their targets and their precise physiologic contributions to retinoid homeostasis and function.
Collapse
Affiliation(s)
- Joseph L Napoli
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, 119 Morgan Hall, 94720, Berkeley, CA, USA.
| |
Collapse
|
79
|
Yang D, Krois CR, Huang P, Wang J, Min J, Yoo HS, Deng Y, Napoli JL. Raldh1 promotes adiposity during adolescence independently of retinal signaling. PLoS One 2017; 12:e0187669. [PMID: 29095919 PMCID: PMC5667840 DOI: 10.1371/journal.pone.0187669] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 10/24/2017] [Indexed: 12/26/2022] Open
Abstract
All-trans-retinoic acid (RA) inhibits adipogenesis in established preadipocyte cell lines. Dosing pharmacological amounts of RA reduces weight gain in mice fed a high-fat diet, i.e. counteracts diet-induced obesity (DIO). The aldehyde dehydrogenase Raldh1 (Aldh1a1) functions as one of three enzymes that converts the retinol metabolite retinal into RA, and one of many proteins that contribute to RA homeostasis. Female Raldh1-ablated mice resist DIO. This phenotype contrasts with ablations of other enzymes and binding-proteins that maintain RA homeostasis, which gain adiposity. The phenotype observed prompted the conclusion that loss of Raldh1 causes an increase in adipose tissue retinal, and therefore, retinal functions independently of RA to prevent DIO. A second deduction proposed that low nM concentrations of RA stimulate adipogenesis, in contrast to higher concentrations. Using peer-reviewed LC/MS/MS assays developed and validated for quantifying tissue RA and retinal, we show that endogenous retinal and RA concentrations in adipose tissues from Raldh1-null mice do not correlate with the phenotype. Moreover, male Raldh1-null mice resist weight gain regardless of dietary fat content. Resistance to weight gain occurs during adolescence in both sexes. We show that RA concentrations as low as 1 nM, i.e. in the sub-physiological range, impair adipogenesis of embryonic fibroblasts from wild-type mice. Embryonic fibroblasts from Raldh1-null mice resist differentiating into adipocytes, but retain ability to generate RA. These fibroblasts remain sensitive to an RA receptor pan-agonist, and are not affected by an RA receptor pan-antagonist. Thus, the data do not support the hypothesis that retinal itself represses weight gain and adipogenesis independently of RA. Instead, the data indicate that Raldh1 functions as a retinal and atRA-independent promoter of adiposity during adolescence, and enhances adiposity through pre-adipocyte cell autonomous actions.
Collapse
Affiliation(s)
- Di Yang
- Graduate Program in Metabolic Biology, Nutritional Sciences and Toxicology, University of California, Berkeley, California, United States of America
| | - Charles R. Krois
- Graduate Program in Metabolic Biology, Nutritional Sciences and Toxicology, University of California, Berkeley, California, United States of America
| | - Priscilla Huang
- Graduate Program in Metabolic Biology, Nutritional Sciences and Toxicology, University of California, Berkeley, California, United States of America
| | - Jinshan Wang
- Graduate Program in Metabolic Biology, Nutritional Sciences and Toxicology, University of California, Berkeley, California, United States of America
| | - Jin Min
- Graduate Program in Metabolic Biology, Nutritional Sciences and Toxicology, University of California, Berkeley, California, United States of America
| | - Hong Sik Yoo
- Graduate Program in Metabolic Biology, Nutritional Sciences and Toxicology, University of California, Berkeley, California, United States of America
| | - Yinghua Deng
- Graduate Program in Metabolic Biology, Nutritional Sciences and Toxicology, University of California, Berkeley, California, United States of America
| | - Joseph L. Napoli
- Graduate Program in Metabolic Biology, Nutritional Sciences and Toxicology, University of California, Berkeley, California, United States of America
- * E-mail:
| |
Collapse
|
80
|
Wang B, Fu X, Liang X, Deavila JM, Wang Z, Zhao L, Tian Q, Zhao J, Gomez NA, Trombetta SC, Zhu MJ, Du M. Retinoic acid induces white adipose tissue browning by increasing adipose vascularity and inducing beige adipogenesis of PDGFRα + adipose progenitors. Cell Discov 2017; 3:17036. [PMID: 29021914 PMCID: PMC5633810 DOI: 10.1038/celldisc.2017.36] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 08/31/2017] [Indexed: 12/28/2022] Open
Abstract
Formation of beige adipocytes within white adipose tissue enhances energy expenditure, which is a promising strategy to reduce obesity and prevent metabolic symptoms. Vitamin A and its bioactive metabolite, retinoic acid (RA), have regulatory roles in lipid metabolism. Here we report that RA induces white adipose tissue browning via activating vascular endothelial growth factor (VEGF) signaling. RA triggered angiogenesis and elicited de novo generation of platelet-derived growth factor receptor α positive (PDGFRα+) adipose precursor cells via VEGFA/VEGFR2 signaling. In addition, RA promoted beige/brown adipocyte formation from capillary networks in vitro. Using PDGFRα tracking mice, we found that the vascular system acted as an adipogenic repository by containing PDGFRα+ progenitors which differentiated into beige adipocytes under RA or VEGF164 treatments. Conditional knockout of VEGF receptors blocked RA-stimulated white adipose tissue browning. Moreover, the VEGFA and RA activated p38MAPK to enhance the binding of RA receptor to RA response elements of the Prdm16 promoter and upregulated Prdm16 transcription. In conclusion, RA induces white adipose tissue browning by increasing adipose vascularity and promoting beige adipogenesis of PDGFRα+ adipose progenitors.
Collapse
Affiliation(s)
- Bo Wang
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA, USA.,Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Xing Fu
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Xingwei Liang
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Jeanene M Deavila
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Zhixiu Wang
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Liang Zhao
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Qiyu Tian
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Junxing Zhao
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Noe Alberto Gomez
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Sophie C Trombetta
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Mei-Jun Zhu
- School of Food Sciences, Washington State University, Pullman, WA, USA
| | - Min Du
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA, USA.,Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| |
Collapse
|
81
|
Roussel AM. Déficits en micronutriments dans le surpoids et l’obésité : conséquences métaboliques et cliniques. NUTR CLIN METAB 2017. [DOI: 10.1016/j.nupar.2017.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
82
|
RNA-seq analysis of the kidneys of broiler chickens fed diets containing different concentrations of calcium. Sci Rep 2017; 7:11740. [PMID: 28924246 PMCID: PMC5603577 DOI: 10.1038/s41598-017-11379-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 08/23/2017] [Indexed: 01/13/2023] Open
Abstract
Calcium (Ca) is required for normal growth and is involved in cellular physiology, signal transduction, and bone mineralization. In humans, inadequate Ca intake causes hypocalcaemia, and excessive Ca intake causes hypercalcemia. In chicken, Ca is also required for body weight gain and eggshell formation. However, transcriptomic responses to low/high Ca intake, and mechanisms affecting body weight have not been explored. In this study, we performed comparative RNA sequencing (RNA-seq) using the kidney of broiler chickens fed diets containing 0.8, 1.0, and 1.2% Ca. Annotation of RNA-seq data revealed a significant number of differentially expressed genes (DEGs) in the kidney via pairwise comparison using Cufflinks and edgeR. Using edgeR, we identified 12 DEGs; seven overlapped with those found by cufflinks. Seven DEGs were validated by real-time quantitative-PCR (qRT-PCR) in Ca-supplemented kidneys, and the results correlated with the RNA-seq data. DEGs identified by cufflinks/edgeR were subjected to pathway enrichment, protein/protein interaction, and co-occurrence analyses to determine their involvement in disease. The National Research Council (NRC) recommended Ca intake for 21-day post-hatch broilers is about 1.0%. Our findings suggest that higher-than-recommended Ca intake (1.2%) could reduce body weight gain in broilers, and that affected DEGs are related to stress-induced diseases, such as hypertension.
Collapse
|
83
|
|
84
|
McGowan SE, McCoy DM. Glucocorticoids Retain Bipotent Fibroblast Progenitors during Alveolar Septation in Mice. Am J Respir Cell Mol Biol 2017; 57:111-120. [PMID: 28530121 DOI: 10.1165/rcmb.2016-0376oc] [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] [Indexed: 12/16/2022] Open
Abstract
Glucocorticoids have been widely used and exert pleiotropic effects on alveolar structure and function, but do not improve the long-term clinical outcomes for patients with bronchopulmonary dysplasia, emphysema, or interstitial lung diseases. Treatments that foster alveolar regeneration could substantially improve the long-term outcomes for such patients. One approach to alveolar regeneration is to stimulate and guide intrinsic alveolar progenitors along developmental pathways used during secondary septation. Other investigators and we have identified platelet-derived growth factor receptor-α-expressing fibroblast subpopulations that are alternatively skewed toward myofibroblast or lipofibroblast phenotypes. In this study, we administered either the glucocorticoid receptor agonist dexamethasone (Dex) or the antagonist mifepristone to mice during the first postnatal week and evaluated their effects on cellular proliferation and adoption of α-smooth muscle actin and lipid droplets (markers of the myofibroblast and lipofibroblast phenotypes, respectively). We observed that Dex increased the relative abundance of fibroblasts with progenitor characteristics, i.e., containing both α-smooth muscle actin and lipid droplets, uncoupling protein-1 (a marker of brown and beige adipocytes), delta-like ligand-1, and stem cell antigen-1. Dex enhanced signaling through the Smad1/5 pathway, which increased uncoupling protein-1 in a lung fibroblast progenitor cell line. We conclude that glucocorticoid receptor manipulation can sustain fibroblast plasticity, and posit that targeting downstream glucocorticoid responsive pathways could steer fibroblast progenitors along more desirable regenerative pathways.
Collapse
Affiliation(s)
- Stephen E McGowan
- Department of Veterans Affairs Research Service and.,Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Diann M McCoy
- Department of Veterans Affairs Research Service and.,Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| |
Collapse
|
85
|
Wang B, Wang Z, de Avila JM, Zhu MJ, Zhang F, Gomez NA, Zhao L, Tian Q, Zhao J, Maricelli J, Zhang H, Rodgers BD, Du M. Moderate alcohol intake induces thermogenic brown/beige adipocyte formation via elevating retinoic acid signaling. FASEB J 2017; 31:4612-4622. [PMID: 28679528 DOI: 10.1096/fj.201700396r] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Accepted: 06/19/2017] [Indexed: 12/16/2022]
Abstract
Clinically, low and moderate alcohol intake improves human health with protection against metabolic syndromes, including type 2 diabetes; however, mechanisms that are associated with these effects remain to be elucidated. The aims of this study were to investigate the effects of moderate alcohol intake on thermogenic brown/beige adipocyte formation and glucose and lipid homeostasis, as well as the involvement of retinoic acid (RA) signaling in the entire process. C57BL6 male mice were supplemented with 8% (w/v) alcohol in water for 1 or 4 mo. Alcohol intake prevented body weight gain, induced the formation of uncoupling protein 1-positive beige adipocytes in white adipose tissue, and increased thermogenesis in mice, which is associated with decreased serum glucose and triacylglycerol levels. Mechanistically, alcohol intake increased RA levels in serum and adipose tissue, which was associated with increased expression of aldehyde dehydrogenase family 1 subfamily A1 (Aldh1a1). When RA receptor-α signaling was conditionally blocked in platelet-derived growth factor receptor-α-positive adipose progenitors, the effects of alcohol on beige adipogenesis were largely abolished. Finally, moderate alcohol prevented high-fat diet-induced obesity and metabolic dysfunction. In conclusion, moderate alcohol intake induces thermogenic brown/beige adipocyte formation and promotes glucose and lipid oxidation via elevation of RA signaling.-Wang, B., Wang, Z., de Avila, J. M., Zhu, M.-J., Zhang, F., Gomez, N. A., Zhao, L., Tian, Q., Zhao, J., Maricelli, J., Zhang, H., Rodgers, B. D., Du, M. Moderate alcohol intake induces thermogenic brown/beige adipocyte formation via elevating retinoic acid signaling.
Collapse
Affiliation(s)
- Bo Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China.,Department of Animal Sciences, Washington State University, Pullman, Washington, USA
| | - Zhixiu Wang
- Department of Animal Sciences, Washington State University, Pullman, Washington, USA
| | - Jeanene M de Avila
- Department of Animal Sciences, Washington State University, Pullman, Washington, USA
| | - Mei-Jun Zhu
- School of Food Science, Washington State University, Pullman, Washington, USA
| | - Faya Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington, USA
| | - Noe Alberto Gomez
- Department of Animal Sciences, Washington State University, Pullman, Washington, USA
| | - Liang Zhao
- Department of Animal Sciences, Washington State University, Pullman, Washington, USA
| | - Qiyu Tian
- Department of Animal Sciences, Washington State University, Pullman, Washington, USA
| | - Junxing Zhao
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Shanxi, China
| | - Joseph Maricelli
- School of Molecular Biosciences, Washington State University, Pullman, Washington, USA
| | - Hui Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington, USA
| | - Buel D Rodgers
- Department of Animal Sciences, Washington State University, Pullman, Washington, USA
| | - Min Du
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China; .,Department of Animal Sciences, Washington State University, Pullman, Washington, USA.,College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Shanxi, China
| |
Collapse
|
86
|
Jiang Y, Berry DC, Jo A, Tang W, Arpke RW, Kyba M, Graff JM. A PPARγ transcriptional cascade directs adipose progenitor cell-niche interaction and niche expansion. Nat Commun 2017. [PMID: 28649987 PMCID: PMC5490270 DOI: 10.1038/ncomms15926] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Adipose progenitor cells (APCs) reside in a vascular niche, located within the perivascular compartment of adipose tissue blood vessels. Yet, the signals and mechanisms that govern adipose vascular niche formation and APC niche interaction are unknown. Here we show that the assembly and maintenance of the adipose vascular niche is controlled by PPARγ acting within APCs. PPARγ triggers a molecular hierarchy that induces vascular sprouting, APC vessel niche affinity and APC vessel occupancy. Mechanistically, PPARγ transcriptionally activates PDGFRβ and VEGF. APC expression and activation of PDGFRβ promotes the recruitment and retention of APCs to the niche. Pharmacologically, targeting PDGFRβ disrupts APC niche contact thus blocking adipose tissue expansion. Moreover, enhanced APC expression of VEGF stimulates endothelial cell proliferation and expands the adipose niche. Consequently, APC niche communication and retention are boosted by VEGF thereby impairing adipogenesis. Our data indicate that APCs direct adipose tissue niche expansion via a PPARγ-initiated PDGFRβ and VEGF transcriptional axis. Adipocyte progenitor cells (APCs) are found tethered to adipose tissue blood vessel walls and can differentiate into adipocytes. Here the authors show that PPARγ controls angiogenesis by stimulating APC–blood vessel interaction and retention via a transcriptional network that includes PDGFRβ and VEGF.
Collapse
Affiliation(s)
- Yuwei Jiang
- Division of Endocrinology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Daniel C Berry
- Division of Endocrinology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Ayoung Jo
- Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Wei Tang
- Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Robert W Arpke
- Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota 55455, USA.,Department of Medicine, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Michael Kyba
- Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota 55455, USA.,Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Jonathan M Graff
- Division of Endocrinology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
87
|
Lindholm-Perry AK, Cunningham HC, Kuehn LA, Vallet JL, Keele JW, Foote AP, Cammack KM, Freetly HC. Relationships between the genes expressed in the mesenteric adipose tissue of beef cattle and feed intake and gain. Anim Genet 2017; 48:386-394. [PMID: 28568315 DOI: 10.1111/age.12565] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2017] [Indexed: 12/26/2022]
Abstract
Mesenteric fat, a depot within the visceral fat, accumulates in cattle during maturation and finishing and may be a potential source of production inefficiency. The aim of this study was to determine whether the genes expressed in the mesenteric fat of steers were associated with body weight gain and feed intake. Sixteen steers chosen by their rank of distance from the bivariate mean for gain and feed intake were used for this study. Mesenteric fat was obtained and evaluated for differences in gene expression. A total of 1831 genes were identified as differentially expressed among steers with variation in feed intake and gain. Many of these genes were involved with metabolic processes such as proteolysis, transcription and translation. In addition, the Gene Ontology annotations including transport and localization were both over-represented among the differentially expressed genes. Pathway analysis was also performed on the differentially expressed genes. The superoxide radical degradation pathway was identified as over-represented based on the differential expression of the genes GPX7, SOD2 and TYRP1, suggesting a potential role for oxidative stress or inflammatory pathways among low gain-high intake animals. GPX7 and SOD2 were in lower transcript abundance, and TYRP1 was higher in transcript abundance among the low gain-high feed intake animals. The retinoate biosynthesis pathway was also enriched due to the differential expression of the genes AKR1C3, ALDH8A1, RDH8, RDH13 and SDR9C7. These genes were all more highly expressed in the low gain-high intake animals. The glycerol degradation and granzyme A signaling pathways were both associated with gain. Three glycerol kinase genes and the GZMA gene were differentially expressed among high vs. low gain animals. Mesenteric fat is a metabolically active tissue, and in this study, genes involved in proteolysis, transcription, translation, transport immune function, glycerol degradation and oxidative stress were differentially expressed among beef steers with variation in body weight gain and feed intake.
Collapse
Affiliation(s)
- A K Lindholm-Perry
- USDA, ARS, U.S. Meat Animal Research Center, P.O. Box 166, Clay Center, NE, 68933, USA
| | - H C Cunningham
- Department of Animal Science, University of Wyoming, Laramie, WY, 82070, USA
| | - L A Kuehn
- USDA, ARS, U.S. Meat Animal Research Center, P.O. Box 166, Clay Center, NE, 68933, USA
| | - J L Vallet
- USDA, ARS, U.S. Meat Animal Research Center, P.O. Box 166, Clay Center, NE, 68933, USA
| | - J W Keele
- USDA, ARS, U.S. Meat Animal Research Center, P.O. Box 166, Clay Center, NE, 68933, USA
| | - A P Foote
- USDA, ARS, U.S. Meat Animal Research Center, P.O. Box 166, Clay Center, NE, 68933, USA
| | - K M Cammack
- Department of Animal Science, University of Wyoming, Laramie, WY, 82070, USA
| | - H C Freetly
- USDA, ARS, U.S. Meat Animal Research Center, P.O. Box 166, Clay Center, NE, 68933, USA
| |
Collapse
|
88
|
Cao J, Ma Y, Yao W, Zhang X, Wu D. Retinoids Regulate Adipogenesis Involving the TGFβ/SMAD and Wnt/β-Catenin Pathways in Human Bone Marrow Mesenchymal Stem Cells. Int J Mol Sci 2017; 18:ijms18040842. [PMID: 28420144 PMCID: PMC5412426 DOI: 10.3390/ijms18040842] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 03/31/2017] [Accepted: 04/06/2017] [Indexed: 12/21/2022] Open
Abstract
Retinoids may regulate cell differentiation as ligands of retinoic acid receptors (RARs) and/or retinoid X receptors (RXRs). We showed that RAR agonists promoted adipogenesis by upregulating the expression of CCAAT/enhancer-binding protein β (C/EBPβ) in the early stages, but blocked adipogenesis at a later stage in human bone marrow mesenchymal stem cells (hBMSCs). RXR agonists promoted adipogenesis at all time points in hBMSCs. The effect of RAR agonists was mediated mainly by the RARβ subtype. RAR agonists, in contrast to RXR agonists, significantly promoted the expression of RARβ. Knockdown of the RARβ gene via small hairpin RNA (shRNA) attenuated the inhibition of RAR agonists toward adipogenesis. Furthermore, we found that RAR agonists upregulated the transforming growth factor β (TGFβ)/SMAD pathway and Wnt/β-catenin pathway on adipogenesis in hBMSCs, and the stimulating effects were noticeably decreased with the RARβ gene knockdown. Both RAR agonists and RXR agonists inhibited adipogenesis and blocked the promoter activity of C/EBPβ and peroxisome proliferator-activated receptor γ (PPARγ) in SW872 cell. These results indicated the RAR agonists perform dual roles in adipogenesis in hBMSCs, and the TGFβ/SMAD pathway and Wnt/β-catenin pathway may involve the inhibitory effect of RAR agonists. RARβ is the main receptor subtype mediating the effect. The roles of RXR agonists in adipogenesis exhibited cell type-specific differences, and may be based on the integration of signals from different RXR dimers.
Collapse
Affiliation(s)
- Jun Cao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China.
| | - Yuhong Ma
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China.
| | - Weiqi Yao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China.
| | - Xiaoye Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China.
| | - Dongcheng Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
89
|
Wang B, Fu X, Liang X, Wang Z, Yang Q, Zou T, Nie W, Zhao J, Gao P, Zhu MJ, de Avila JM, Maricelli J, Rodgers BD, Du M. Maternal Retinoids Increase PDGFRα + Progenitor Population and Beige Adipogenesis in Progeny by Stimulating Vascular Development. EBioMedicine 2017; 18:288-299. [PMID: 28408241 PMCID: PMC5405191 DOI: 10.1016/j.ebiom.2017.03.041] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 03/18/2017] [Accepted: 03/27/2017] [Indexed: 12/31/2022] Open
Abstract
Maternal vitamin A intake varies but its impact on offspring metabolic health is unknown. Here we found that maternal vitamin A or retinoic acid (RA) administration expanded PDGFRα+ adipose progenitor population in progeny, accompanied by increased blood vessel density and enhanced brown-like (beige) phenotype in adipose tissue, protecting offspring from obesity. Blockage of retinoic acid signaling by either BMS493 or negative RA receptor (RARαDN) over-expression abolished the increase in blood vessel density, adipose progenitor population, and beige adipogenesis stimulated by RA. Furthermore, RA-induced beige adipogenesis was blocked following vascular endothelial growth factor receptor (VEGFR) 2 knock out in PDGFRα+ cells, suggesting its mediatory role. Our data reveal an intrinsic link between maternal retinoid level and offspring health via promoting beige adipogenesis. Thus, enhancing maternal retinoids is an amiable therapeutic strategy to prevent obesity in offspring, especially for those born to obese mothers which account for one third of all pregnancies. Maternal vitamin A supplementation increases blood vessel density and expands adipose progenitor population in progeny. Maternal vitamin A supplementation enhances brown-like phenotype in adipose tissues. Maternal vitamin A supplementation protects offspring from diet induced obesity.
Vitamin A and its metabolite, retinoic acid, play key roles in adipogenesis and energy expenditure of adipose tissues. In mice and humans, vitamin A intake is inversely correlated with adiposity. This study has uncovered a role for maternal retinoids in fetal adipose development. Maternal vitamin A supplementation or RA administration increases adipose progenitor population and promotes beige adipogenesis, which protects offspring from diet induced obesity in later life.
Collapse
Affiliation(s)
- Bo Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, 100194, China; Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States
| | - Xing Fu
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States
| | - Xingwei Liang
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States
| | - Zhixiu Wang
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States
| | - Qiyuan Yang
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States
| | - Tiande Zou
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States
| | - Wei Nie
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States
| | - Junxing Zhao
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States; College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Pengfei Gao
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States; College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Mei-Jun Zhu
- School of Food Sciences, Washington State University, Pullman, WA, 99164, United States
| | - Jeanene M de Avila
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States
| | - Joseph Maricelli
- School of Molecular Biosciences, Washington State University, Pullman, WA 99164, United States
| | - Buel D Rodgers
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States
| | - Min Du
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, 100194, China; Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States; College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China.
| |
Collapse
|
90
|
Inhibition of fat cell differentiation in 3T3-L1 pre-adipocytes by all-trans retinoic acid: Integrative analysis of transcriptomic and phenotypic data. BIOMOLECULAR DETECTION AND QUANTIFICATION 2016; 11:31-44. [PMID: 28331816 PMCID: PMC5348118 DOI: 10.1016/j.bdq.2016.11.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 11/08/2016] [Accepted: 11/15/2016] [Indexed: 01/10/2023]
Abstract
The process of adipogenesis is controlled in a highly orchestrated manner, including transcriptional and post-transcriptional events. In developing 3T3-L1 pre-adipocytes, this program can be interrupted by all-trans retinoic acid (ATRA). To examine this inhibiting impact by ATRA, we generated large-scale transcriptomic data on the microRNA and mRNA level. Non-coding RNAs such as microRNAs represent a field in RNA turnover, which is very important for understanding the regulation of mRNA gene expression. High throughput mRNA and microRNA expression profiling was performed using mRNA hybridisation microarray technology and multiplexed expression assay for microRNA quantification. After quantitative measurements we merged expression data sets, integrated the results and analysed the molecular regulation of in vitro adipogenesis. For this purpose, we applied local enrichment analysis on the integrative microRNA-mRNA network determined by a linear regression approach. This approach includes the target predictions of TargetScan Mouse 5.2 and 23 pre-selected, significantly regulated microRNAs as well as Affymetrix microarray mRNA data. We found that the cellular lipid metabolism is negatively affected by ATRA. Furthermore, we were able to show that microRNA 27a and/or microRNA 96 are important regulators of gap junction signalling, the rearrangement of the actin cytoskeleton as well as the citric acid cycle, which represent the most affected pathways with regard to inhibitory effects of ATRA in 3T3-L1 preadipocytes. In conclusion, the experimental workflow and the integrative microRNA–mRNA data analysis shown in this study represent a possibility for illustrating interactions in highly orchestrated biological processes. Further the applied global microRNA–mRNA interaction network may also be used for the pre-selection of potential new biomarkers with regard to obesity or for the identification of new pharmaceutical targets.
Collapse
|
91
|
Libien J, Kupersmith MJ, Blaner W, McDermott MP, Gao S, Liu Y, Corbett J, Wall M. Role of vitamin A metabolism in IIH: Results from the idiopathic intracranial hypertension treatment trial. J Neurol Sci 2016; 372:78-84. [PMID: 28017254 DOI: 10.1016/j.jns.2016.11.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 11/05/2016] [Accepted: 11/08/2016] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Vitamin A and its metabolites (called retinoids) have been thought to play a role in the development of idiopathic intracranial hypertension (IIH). The IIH Treatment Trial (IIHTT) showed the efficacy of acetazolamide (ACZ) in improving visual field function, papilledema grade, quality of life and cerebrospinal fluid (CSF) pressure. We postulated that IIH patients would demonstrate elevated measures of vitamin A metabolites in the serum and CSF. METHODS Comprehensive measures of serum vitamin A and its metabolites were obtained from 96 IIHTT subjects, randomly assigned to treatment with ACZ or placebo, and 25 controls with similar gender, age and body mass index (BMI). These included retinol, retinol binding protein, all-trans retinoic acid (ATRA), alpha- and beta-carotenes, and beta-cryptoxanthin. The IIHTT subjects also had CSF and serum vitamin A and metabolite measurements obtained at study entry and at six months. RESULTS At study entry, of the vitamin A metabolites only serum ATRA was significantly different in IIHTT subjects (median 4.33nM) and controls (median 5.04nM, p=0.02). The BMI of IIHTT subjects showed mild significant negative correlations with serum ATRA, alpha- and beta-carotene, and beta-cryptoxanthin. In contrast, the control subject BMI correlated only with serum ATRA. At six months, the serum retinol, alpha-carotene, beta-carotene, and CSF retinol were increased from baseline in the ACZ treated group, but only increases in alpha-carotene (p=0.02) and CSF ATRA (p=0.04) were significantly greater in the ACZ group compared with the placebo group. No other vitamin A measures were significantly altered over the six months in either treatment group. Weight loss correlated with only with the change in serum beta-carotene (r=-0.44, p=0.006) and the change in CSF retinol (r=-0.61, p=0.02). CONCLUSION Vitamin A toxicity is unlikely a contributory factor in the causation of IIH. Our findings differ from those of prior reports in part because of our use of more accurate quantitative methods and measuring vitamin A metabolites in both serum and CSF. ACZ may alter retinoid metabolism in IIH patients.
Collapse
Affiliation(s)
- J Libien
- Pathology, State University of New York, Downstate School of Medicine, Brooklyn, NY, United States
| | - M J Kupersmith
- Neurology and Ophthalmology, Mount Sinai West, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| | - W Blaner
- Medicine, College of Physicians and Surgeons, Columbia University School of Medicine, New York, NY, United States
| | - M P McDermott
- Biostatistics, University of Rochester, Rochester, NY, United States
| | - S Gao
- Biostatistics, University of Rochester, Rochester, NY, United States
| | - Y Liu
- Pathology, State University of New York, Downstate School of Medicine, Brooklyn, NY, United States
| | - J Corbett
- Neurology, University of Mississippi School of Medicine, Jackson, MS, United States
| | - M Wall
- Neurology, University of Iowa School of Medicine, Iowa City, IA, United States
| | | |
Collapse
|
92
|
Parameswaran S, Sharma RK. Insulin Cannot Induce Adipogenic Differentiation in Primary Cardiac Cultures. Int J Angiol 2016; 25:181-5. [PMID: 27574386 DOI: 10.1055/s-0035-1571191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 11/30/2015] [Indexed: 10/22/2022] Open
Abstract
Cardiac tissue contains a heterogeneous population of cardiomyocytes and nonmyocyte population especially fibroblasts. Fibroblast differentiation into adipogenic lineage is important for fat accumulation around the heart which is important in cardiac pathology. The differentiation in fibroblast has been observed both spontaneously and due to increased insulin stimulation. The present study aims to observe the effect of insulin in adipogenic differentiation of cardiac cells present in primary murine cardiomyocyte cultures. Oil Red O (ORO) staining has been used for observing the lipid accumulations formed due to adipogenic differentiation in murine cardiomyocyte cultures. The accumulated lipids were quantified by ORO assay and normalized using protein estimation. The lipid accumulation in cardiac cultures did not increase in presence of insulin. However, addition of other growth factors like insulin-like growth factor 1 and epidermal growth factor promoted adipogenic differentiation even in the presence of insulin and other inhibitory molecules such as vitamins. Lipid accumulation also increased in cells grown in media without insulin after an initial exposure to insulin-containing growth media. The current study adds to the existing knowledge that the insulin by itself cannot induce adipogenic induction in the cardiac cultures. The data have significance in the understanding of cardiovascular health especially in diabetic patients.
Collapse
Affiliation(s)
- Sreejit Parameswaran
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Rajendra K Sharma
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
93
|
Berry DC, Jiang Y, Graff JM. Emerging Roles of Adipose Progenitor Cells in Tissue Development, Homeostasis, Expansion and Thermogenesis. Trends Endocrinol Metab 2016; 27:574-585. [PMID: 27262681 PMCID: PMC10947416 DOI: 10.1016/j.tem.2016.05.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 04/26/2016] [Accepted: 05/02/2016] [Indexed: 01/10/2023]
Abstract
Stem or progenitor cells are an essential component for the development, homeostasis, expansion, and regeneration of many tissues. Within white adipose tissue (WAT) reside vascular-resident adipose progenitor cells (APCs) that can proliferate and differentiate into either white or beige/brite adipocytes, which may control adiposity. Recent studies have begun to show that APCs can be manipulated to control adiposity and counteract 'diabesity'. However, much remains unknown about the identity of APCs and how they may control adiposity in response to homeostatic and external cues. Here, we discuss recent advances in our understanding of adipose progenitors and cover a range of topics, including the stem cell/progenitor lineage, their niche, their developmental and adult roles, and their role in cold-induced beige/brite adipocyte formation.
Collapse
Affiliation(s)
- Daniel C Berry
- Division of Endocrinology, Department of Internal Medicine, University of Texas Southwestern Medical Center 5323, Harry Hines Blvd, Dallas, TX 75235, USA
| | - Yuwei Jiang
- Division of Endocrinology, Department of Internal Medicine, University of Texas Southwestern Medical Center 5323, Harry Hines Blvd, Dallas, TX 75235, USA
| | - Jonathan M Graff
- Division of Endocrinology, Department of Internal Medicine, University of Texas Southwestern Medical Center 5323, Harry Hines Blvd, Dallas, TX 75235, USA; Department of Molecular Biology, University of Texas Southwestern Medical Center 5323, Harry Hines Blvd, Dallas, TX 75235, USA.
| |
Collapse
|
94
|
Whole Blood RNA as a Source of Transcript-Based Nutrition- and Metabolic Health-Related Biomarkers. PLoS One 2016; 11:e0155361. [PMID: 27163124 PMCID: PMC4862680 DOI: 10.1371/journal.pone.0155361] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 04/27/2016] [Indexed: 12/17/2022] Open
Abstract
Blood cells are receiving an increasing attention as an easily accessible source of transcript-based biomarkers. We studied the feasibility of using mouse whole blood RNA in this context. Several paradigms were studied: (i) metabolism-related transcripts known to be affected in rat tissues and peripheral blood mononuclear cells (PBMC) by fasting and upon the development of high fat diet (HFD)-induced overweight were assessed in whole blood RNA of fasted rats and mice and of HFD-fed mice; (ii) retinoic acid (RA)-responsive genes in tissues were assessed in whole blood RNA of control and RA-treated mice; (iii) lipid metabolism-related transcripts previously identified in PBMC as potential biomarkers of metabolic health in a rat model were assessed in whole blood in an independent model, namely retinoblastoma haploinsufficient (Rb+/-) mice. Blood was collected and stored in RNAlater® at -80°C until analysis of selected transcripts by real-time RT-PCR. Comparable changes with fasting were detected in the expression of lipid metabolism-related genes when RNA from either PBMC or whole blood of rats or mice was used. HFD-induced excess body weight and fat mass associated with expected changes in the expression of metabolism-related genes in whole blood of mice. Changes in gene expression in whole blood of RA-treated mice reproduced known transcriptional actions of RA in hepatocytes and adipocytes. Reduced expression of Fasn, Lrp1, Rxrb and Sorl1 could be validated as early biomarkers of metabolic health in young Rb+/- mice using whole blood RNA. Altogether, these results support the use of whole blood RNA in studies aimed at identifying blood transcript-based biomarkers of nutritional/metabolic status or metabolic health. Results also support reduced expression of Fasn, Lrp1, Rxrb and Sorl1 in blood cells at young age as potential biomarkers of metabolic robustness.
Collapse
|
95
|
Mogi M, Kohara K, Nakaoka H, Kan-No H, Tsukuda K, Wang XL, Chisaka T, Bai HY, Shan BS, Kukida M, Iwanami J, Miki T, Horiuchi M. Diabetic mice exhibited a peculiar alteration in body composition with exaggerated ectopic fat deposition after muscle injury due to anomalous cell differentiation. J Cachexia Sarcopenia Muscle 2016; 7:213-24. [PMID: 27493874 PMCID: PMC4864245 DOI: 10.1002/jcsm.12044] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 04/04/2015] [Accepted: 04/23/2015] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Sarcopenic obesity, age-related muscle loss, which is compensated by an increase in fat mass, impairs quality of life in elderly people. Although the increase in intramuscular fat is associated with decreased insulin sensitivity and increased metabolic risk factors, the origin of diabetes-associated intramuscular fat has not been elucidated. Here, we investigated intramuscular fat deposition using a muscle injury model in type 2 diabetic mice. METHODS Male 8-week-old C57BL/6 and 8-week-old and 26-week-old KKAy underwent intramuscular injection of cardiotoxin (Ctx) (100 μL/10 μM) into the tibialis anterior (TA) muscles. After 2 weeks, the muscles were removed and evaluated. RESULTS KKAy exhibited impaired muscle regeneration and ectopic fat deposition. Such impairment was more marked in older KKAy. These changes were also observed in another diabetic mouse model, db/db and diet-induced obese mice but not in streptozocin-induced diabetic mice. Deposited fat was platelet-derived growth factor (PDGF) receptor alpha positive and its cytoskeleton was stained with Masson's trichrome, indicating it to be of fibro-adipocyte progenitor cell origin. Expression of a myogenic marker, myoD, was lower and that of PDGF receptor alpha and CCAAT/enhancer binding protein (CEBP) alpha was higher in Ctx-injured TA of KKAy compared with that of C57BL/6. Peroxisome proliferator-activated receptor γ (PPARγ) was highly expressed in fat-forming lesions in older KKAy. Treatment with all-trans retinoic acid prevented the formation of intramuscular fat; however, treatment with GW9662, a PPARγ antagonist, increased the fibrotic change in muscle. CONCLUSIONS Diabetic mice showed impaired muscle regeneration with fat deposition, suggesting that diabetes may enhance sarcopenic obesity through a mechanism involving anomalous fibro-adipocyte progenitor cell differentiation.
Collapse
Affiliation(s)
- Masaki Mogi
- Department of Molecular Cardiovascular Biology and Pharmacology Ehime University, Graduate School of Medicine Tohon Ehime 791-0295 Japan
| | - Katsuhiko Kohara
- Department of Neurology and Geriatric Medicine Ehime University, Graduate School of Medicine Tohon Ehime 791-0295 Japan
| | - Hirotomo Nakaoka
- Department of Molecular Cardiovascular Biology and Pharmacology Ehime University, Graduate School of Medicine Tohon Ehime 791-0295 Japan
| | - Harumi Kan-No
- Department of Molecular Cardiovascular Biology and Pharmacology Ehime University, Graduate School of Medicine Tohon Ehime 791-0295 Japan
| | - Kana Tsukuda
- Department of Molecular Cardiovascular Biology and Pharmacology Ehime University, Graduate School of Medicine Tohon Ehime 791-0295 Japan
| | - Xiao-Li Wang
- Department of Molecular Cardiovascular Biology and Pharmacology Ehime University, Graduate School of Medicine Tohon Ehime 791-0295 Japan
| | - Toshiyuki Chisaka
- Department of Molecular Cardiovascular Biology and Pharmacology Ehime University, Graduate School of Medicine Tohon Ehime 791-0295 Japan; Department of Pediatrics Ehime University, Graduate School of Medicine Tohon Ehime 791-0295 Japan
| | - Hui-Yu Bai
- Department of Molecular Cardiovascular Biology and Pharmacology Ehime University, Graduate School of Medicine Tohon Ehime 791-0295 Japan
| | - Bao-Shuai Shan
- Department of Molecular Cardiovascular Biology and Pharmacology Ehime University, Graduate School of Medicine Tohon Ehime 791-0295 Japan
| | - Masayoshi Kukida
- Department of Molecular Cardiovascular Biology and Pharmacology Ehime University, Graduate School of Medicine Tohon Ehime 791-0295 Japan; Department of Cardiology, Pulmonology, Hypertension and Nephrology Ehime University, Graduate School of Medicine Tohon Ehime 791-0295 Japan
| | - Jun Iwanami
- Department of Molecular Cardiovascular Biology and Pharmacology Ehime University, Graduate School of Medicine Tohon Ehime 791-0295 Japan
| | - Tetsuro Miki
- Department of Neurology and Geriatric Medicine Ehime University, Graduate School of Medicine Tohon Ehime 791-0295 Japan
| | - Masatsugu Horiuchi
- Department of Molecular Cardiovascular Biology and Pharmacology Ehime University, Graduate School of Medicine Tohon Ehime 791-0295 Japan
| |
Collapse
|
96
|
Wang B, Yang Q, Harris CL, Nelson ML, Busboom JR, Zhu MJ, Du M. Nutrigenomic regulation of adipose tissue development - role of retinoic acid: A review. Meat Sci 2016; 120:100-106. [PMID: 27086067 DOI: 10.1016/j.meatsci.2016.04.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 04/02/2016] [Accepted: 04/05/2016] [Indexed: 12/17/2022]
Abstract
To improve the efficiency of animal production, livestock have been extensively selected or managed to reduce fat accumulation and increase lean growth, which reduces intramuscular or marbling fat content. To enhance marbling, a better understanding of the mechanisms regulating adipogenesis is needed. Vitamin A has recently been shown to have a profound impact on all stages of adipogenesis. Retinoic acid, an active metabolite of vitamin A, activates both retinoic acid receptors (RAR) and retinoid X receptors (RXR), inducing epigenetic changes in key regulatory genes governing adipogenesis. Additionally, Vitamin D and folates interact with the retinoic acid receptors to regulate adipogenesis. In this review, we discuss nutritional regulation of adipogenesis, focusing on retinoic acid and its impact on epigenetic modifications of key adipogenic genes.
Collapse
Affiliation(s)
- Bo Wang
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States
| | - Qiyuan Yang
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States
| | - Corrine L Harris
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States
| | - Mark L Nelson
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States
| | - Jan R Busboom
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States
| | - Mei-Jun Zhu
- School of Food Science, Washington State University, Pullman, WA 99164, United States
| | - Min Du
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States.
| |
Collapse
|
97
|
Retinoids have therapeutic action in type 2 diabetes. Nutrition 2016; 32:898-903. [PMID: 27134203 DOI: 10.1016/j.nut.2016.02.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 12/22/2015] [Accepted: 02/02/2016] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Uncontrolled diabetes is associated with a compromised antioxidant state of the body. Consequentially, the reactive oxygen species generated lead to oxidative insult and associated complications. Based on this paradigm, exogenous antioxidant supplementation is thought to exert a therapeutic role in type 2 diabetes (T2-D) biology. METHODS In the present study, the effect of vitamin A supplementation was assessed on disease progression in T2-D BALB/c mice. Animals were divided into three groups. With the exception of control, the mice in remaining groups were induced with experimental T2-D. After a 15-day treatment protocol, the mice were sacrificed and various parameters were analyzed. RESULTS The treated group evidenced a considerable improvement in total antioxidant potential and glycemic control. A therapeutic effect on beta cell degeneration as compared to the diabetic group was also found. CONCLUSIONS The study illustrates the antihyperglycemic and antioxidant potential of vitamin A in vivo, which has potential to serve as a dietary intervention in T2-D.
Collapse
|
98
|
Rahman N, Jeon M, Song HY, Kim YS. Cryptotanshinone, a compound of Salvia miltiorrhiza inhibits pre-adipocytes differentiation by regulation of adipogenesis-related genes expression via STAT3 signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2016; 23:58-67. [PMID: 26902408 DOI: 10.1016/j.phymed.2015.12.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 12/01/2015] [Accepted: 12/07/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUND Cryptotanshinone (CT), a major tanshinone found in Salvia miltiorrhiza Bunge (Lamiaceae), has various pharmacological effects such as antitumor, anti-inflammatory, and antioxidant properties. Despite its well-documented benefits in a wide range of diseases, the effect of CT on adipocyte differentiation has not been well characterized. PURPOSE The present study was designed to determine the in vitro anti-adipogenic effect and underlying molecular mechanisms of CT using 3T3-L1 murine pre-adipocytes. METHODS We measured the levels of intracellular triglyceride accumulation and mRNA and protein expression of key adipogenic transcription factors and their target genes. RESULTS Treatment with CT drastically reduced lipid accumulation in a dose- and time-dependent manner. Molecular assays showed that CT effectively suppressed the expression of C/EBPβ, C/EBPα, and PPARγ and of their target adipocyte-specific genes aP2, adiponectin, and GLUT4 but activated the expression of anti-adipogenic genes such as GATA2, CHOP10, and TNF-α. CT treatment also inhibited the phosphorylation of STAT3 in the early phase of adipogenesis. A small-interfering-RNA-mediated knock-down of STAT3 potentiated the anti-adipogenic effect of CT. CONCLUSION Taken together, the results suggest that CT may be a good anti-adipogenic candidate because it regulates STAT3 during early adipogenesis.
Collapse
Affiliation(s)
- Naimur Rahman
- Department of Microbiology, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Miso Jeon
- Department of Microbiology, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Ho-Yeon Song
- Department of Microbiology, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Yong-Sik Kim
- Department of Microbiology, College of Medicine, Soonchunhyang University, Cheonan, Korea; Institute of Tissue Engineering, College of Medicine, Soonchunhyang University, Cheonan, Korea.
| |
Collapse
|
99
|
Abstract
Cell, animal and human studies dealing with carotenoids and carotenoid derivatives as nutritional regulators of adipose tissue biology with implications for the etiology and management of obesity and obesity-related metabolic diseases are reviewed. Most studied carotenoids in this context are β-carotene, cryptoxanthin, astaxanthin and fucoxanthin, together with β-carotene-derived retinoids and some other apocarotenoids. Studies indicate an impact of these compounds on essential aspects of adipose tissue biology including the control of adipocyte differentiation (adipogenesis), adipocyte metabolism, oxidative stress and the production of adipose tissue-derived regulatory signals and inflammatory mediators. Specific carotenoids and carotenoid derivatives restrain adipogenesis and adipocyte hypertrophy while enhancing fat oxidation and energy dissipation in brown and white adipocytes, and counteract obesity in animal models. Intake, blood levels and adipocyte content of carotenoids are reduced in human obesity. Specifically designed human intervention studies in the field, though still sparse, indicate a beneficial effect of carotenoid supplementation in the accrual of abdominal adiposity. In summary, studies support a role of specific carotenoids and carotenoid derivatives in the prevention of excess adiposity, and suggest that carotenoid requirements may be dependent on body composition.
Collapse
Affiliation(s)
- M Luisa Bonet
- Group of Nutrigenomics and Obesity, Laboratory of Molecular Biology, Nutrition and Biotechnology, Universitat de les Illes Balears, Cra. Valldemossa Km 7.5. 07122, Palma de Mallorca, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Palma de Mallorca, Spain.
| | - Jose A Canas
- Metabolism and Diabetes, Nemours Children's Clinic, Jacksonville, FL, 32207, USA
| | - Joan Ribot
- Group of Nutrigenomics and Obesity, Laboratory of Molecular Biology, Nutrition and Biotechnology, Universitat de les Illes Balears, Cra. Valldemossa Km 7.5. 07122, Palma de Mallorca, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Palma de Mallorca, Spain
| | - Andreu Palou
- Group of Nutrigenomics and Obesity, Laboratory of Molecular Biology, Nutrition and Biotechnology, Universitat de les Illes Balears, Cra. Valldemossa Km 7.5. 07122, Palma de Mallorca, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Palma de Mallorca, Spain
| |
Collapse
|
100
|
Abstract
It has long been established that the transcriptional activity of retinoic acid (RA) is mediated by members of the nuclear receptor family of ligand-activated transcription factors termed RA receptors (RARs). More recent observations have established that RA also activates an additional nuclear receptor, PPARβ/δ. Partitioning RA between RARs and PPARβ/δ is governed by different intracellular lipid-binding proteins: cellular RA binding protein 2 (CRABP2) delivers RA to nuclear RARs and a fatty acid binding protein (FABP5) delivers the hormone from the cytosol to nuclear PPARβ/δ. Consequently, RA signals through RARs in CRABP2-expressing cells, but activates PPARβ/δ in cells that express a high level of FABP5. RA elicits different and sometimes opposing responses in cells that express different FABP5/CRABP2 ratios because PPARβ/δ and RARs regulate the expression of distinct sets of genes. An overview of the observations that led to the discovery of this non-classical activity of RA are presented here, along with a discussion of evidence demonstrating the involvement of the dual transcriptional activities of RA in regulating energy homeostasis, insulin responses, and adipocyte and neuron differentiation.
Collapse
|