1
|
Karimzadeh K, Uju C, Zahmatkesh A, Unniappan S. Fat mass and obesity associated gene and homeobox transcription factor iriquois-3 mRNA profiles in the metabolic tissues of zebrafish are modulated by feeding and food deprivation. Gen Comp Endocrinol 2025; 360:114621. [PMID: 39414090 DOI: 10.1016/j.ygcen.2024.114621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 09/26/2024] [Accepted: 10/07/2024] [Indexed: 10/18/2024]
Abstract
Fat mass and obesity associated gene (FTO) has been strongly associated with obesity, and it is functionally linked to the homeobox transcription factor iriquois-3 (IRX3). In mammals, FTO and IRX3 are involved in the regulation of food intake and metabolism. This study aimed to determine whether FTO and IRX3are affected by feeding and food unavailability. FTO and IRX3 mRNA and protein were found widely distributed in all tissues examined, including the brain, muscle, gut, and liver. Postprandial increase in the abundance of FTO and IRX3 mRNAs was observed in metabolic tissues of both male and female zebrafish at 1 h post-feeding. Meanwhile, their expression in the brain and gut decreased at 3 h post-feeding, reaching preprandial levels. Additionally, FTO and IRX3 mRNA abundance in examined tissues increased after 7 days of food deprivation, but substantially decreased after refeeding for 24 h. In summary, we report that both FTO and IRX3 are meal-sensitive genes in zebrafish. The fasting-induced increase suggests a possible appetite regulatory role for FTO and IRX3 in zebrafish. These findings highlight the importance of FTO and IRX3 in appetite and metabolic regulation in zebrafish.
Collapse
Affiliation(s)
- Katayoon Karimzadeh
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5B4, Canada; Marine Biology Department, Islamic Azad University, Lahijan Branch, Lahijan, Iran
| | - Chinelo Uju
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5B4, Canada
| | - Asgar Zahmatkesh
- Aquaculture Department, Gilan Agricultural and Natural Resources Research and Education Center, AREEO, Gilan, Iran
| | - Suraj Unniappan
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5B4, Canada.
| |
Collapse
|
2
|
Melnik BC, Weiskirchen R, Stremmel W, John SM, Schmitz G. Risk of Fat Mass- and Obesity-Associated Gene-Dependent Obesogenic Programming by Formula Feeding Compared to Breastfeeding. Nutrients 2024; 16:2451. [PMID: 39125332 PMCID: PMC11314333 DOI: 10.3390/nu16152451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/21/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
It is the purpose of this review to compare differences in postnatal epigenetic programming at the level of DNA and RNA methylation and later obesity risk between infants receiving artificial formula feeding (FF) in contrast to natural breastfeeding (BF). FF bears the risk of aberrant epigenetic programming at the level of DNA methylation and enhances the expression of the RNA demethylase fat mass- and obesity-associated gene (FTO), pointing to further deviations in the RNA methylome. Based on a literature search through Web of Science, Google Scholar, and PubMed databases concerning the dietary and epigenetic factors influencing FTO gene and FTO protein expression and FTO activity, FTO's impact on postnatal adipogenic programming was investigated. Accumulated translational evidence underscores that total protein intake as well as tryptophan, kynurenine, branched-chain amino acids, milk exosomal miRNAs, NADP, and NADPH are crucial regulators modifying FTO gene expression and FTO activity. Increased FTO-mTORC1-S6K1 signaling may epigenetically suppress the WNT/β-catenin pathway, enhancing adipocyte precursor cell proliferation and adipogenesis. Formula-induced FTO-dependent alterations of the N6-methyladenosine (m6A) RNA methylome may represent novel unfavorable molecular events in the postnatal development of adipogenesis and obesity, necessitating further investigations. BF provides physiological epigenetic DNA and RNA regulation, a compelling reason to rely on BF.
Collapse
Affiliation(s)
- Bodo C. Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49076 Osnabrück, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, D-52074 Aachen, Germany;
| | - Wolfgang Stremmel
- Praxis for Internal Medicine, Beethovenstrasse 2, D-76530 Baden-Baden, Germany;
| | - Swen Malte John
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49076 Osnabrück, Germany
- Institute for Interdisciplinary Dermatological Prevention and Rehabilitation (iDerm), University of Osnabrück, D-49076 Osnabrück, Germany;
| | - Gerd Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital of Regensburg, D-93053 Regensburg, Germany;
| |
Collapse
|
3
|
Yau-Qiu ZX, Galmés S, Castillo P, Picó C, Palou A, Rodríguez AM. Maternal choline supplementation mitigates premature foetal weight gain induced by an obesogenic diet, potentially linked to increased amniotic fluid leptin levels in rats. Sci Rep 2024; 14:11366. [PMID: 38762543 PMCID: PMC11102553 DOI: 10.1038/s41598-024-62229-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 05/15/2024] [Indexed: 05/20/2024] Open
Abstract
Placental leptin may impact foetal development. Maternal overnutrition has been linked to increased plasma leptin levels and adverse effects on offspring, whereas choline, an essential nutrient for foetal development, has shown promise in mitigating some negative impacts of maternal obesity. Here, we investigate whether a maternal obesogenic diet alters foetal growth and leptin levels in the foetal stomach, amniotic fluid (AF), and placenta in late gestation and explore the potential modulating effects of maternal choline supplementation. Female rats were fed a control (CD) or a western diet (WD) four weeks before mating and during gestation, half of them supplemented with choline (pregnancy days 11-17). Leptin levels (in foetal stomach, AF, and placenta) and leptin gene expression (in placenta) were assessed on gestation days 20 and 21. At day 20, maternal WD feeding resulted in greater leptin levels in foetal stomach, placenta, and AF. The increased AF leptin levels were associated with a premature increase in foetal weight in both sexes. Maternal choline supplementation partially prevented these alterations, but effects differed in CD dams, causing increased AF leptin levels and greater weight in male foetuses at day 20. Maternal choline supplementation effectively mitigates premature foetal overgrowth induced by an obesogenic diet, potentially linked to increased AF leptin levels. Further research is needed to explore the sex-specific effects.
Collapse
Affiliation(s)
- Zhi Xin Yau-Qiu
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation-NuBE), University of the Balearic Islands (UIB), Cra. Valldemossa Km 7.5, 07122, Palma, Balearic Islands, Spain
- Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Spain
- CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Sebastià Galmés
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation-NuBE), University of the Balearic Islands (UIB), Cra. Valldemossa Km 7.5, 07122, Palma, Balearic Islands, Spain.
- Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Spain.
- CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain.
| | - Pedro Castillo
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation-NuBE), University of the Balearic Islands (UIB), Cra. Valldemossa Km 7.5, 07122, Palma, Balearic Islands, Spain
- Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Spain
- CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Catalina Picó
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation-NuBE), University of the Balearic Islands (UIB), Cra. Valldemossa Km 7.5, 07122, Palma, Balearic Islands, Spain
- Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Spain
- CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Andreu Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation-NuBE), University of the Balearic Islands (UIB), Cra. Valldemossa Km 7.5, 07122, Palma, Balearic Islands, Spain
- Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Spain
- CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Ana María Rodríguez
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation-NuBE), University of the Balearic Islands (UIB), Cra. Valldemossa Km 7.5, 07122, Palma, Balearic Islands, Spain
- Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Spain
- CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| |
Collapse
|
4
|
Ferreira SRG, Macotela Y, Velloso LA, Mori MA. Determinants of obesity in Latin America. Nat Metab 2024; 6:409-432. [PMID: 38438626 DOI: 10.1038/s42255-024-00977-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 01/04/2024] [Indexed: 03/06/2024]
Abstract
Obesity rates are increasing almost everywhere in the world, although the pace and timing for this increase differ when populations from developed and developing countries are compared. The sharp and more recent increase in obesity rates in many Latin American countries is an example of that and results from regional characteristics that emerge from interactions between multiple factors. Aware of the complexity of enumerating these factors, we highlight eight main determinants (the physical environment, food exposure, economic and political interest, social inequity, limited access to scientific knowledge, culture, contextual behaviour and genetics) and discuss how they impact obesity rates in Latin American countries. We propose that initiatives aimed at understanding obesity and hampering obesity growth in Latin America should involve multidisciplinary, global approaches that consider these determinants to build more effective public policy and strategies, accounting for regional differences and disease complexity at the individual and systemic levels.
Collapse
Affiliation(s)
| | - Yazmín Macotela
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, UNAM Campus-Juriquilla, Querétaro, Mexico
| | - Licio A Velloso
- Obesity and Comorbidities Research Center, Faculty of Medical Sciences, Universidade Estadual de Campinas, Campinas, Brazil
| | - Marcelo A Mori
- Institute of Biology, Universidade Estadual de Campinas, Campinas, Brazil.
| |
Collapse
|
5
|
Li C, Xu P, Huang Y, Wang Y, Wu Y, Li H, Peng Y, Wang X, Li G, He J, Miao C. RNA methylations in depression, from pathological mechanism to therapeutic potential. Biochem Pharmacol 2023; 215:115750. [PMID: 37595670 DOI: 10.1016/j.bcp.2023.115750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 08/13/2023] [Accepted: 08/15/2023] [Indexed: 08/20/2023]
Abstract
Depression is caused by a variety of factors such as genetic factors, biological factors, and psychosocial factors, and the pathogenesis is complex. RNA methylations and related downstream signaling pathways influence a variety of biological mechanisms, including cell differentiation, tumorigenesis, sex determination, and stress response. In this work, we searched the PubMed, Web of Science, National Library of Science and Technology (NSTL), and ScienceDirect Online (SDOL) databases to summarize the biological roles of RNA methylations and their impact on the pathological mechanisms of depression. RNA methylations play a key role in the development of many diseases, and current research shows that RNA methylations are also closely linked to depression. RNA methylations in depression mainly involve "writers" (mediating the methylation modification process of RNAs), "erasers" (mediating the demethylation modification process of RNA methylation). Fat Mass and Obesity Associated (FTO) influences the development of depression by increasing body mass index (BMI), decreases the dopamine level, inhibits the adrenoceptor beta 2 (ADRB2)-c-Myc-sirt1 pathway, results in the m6A/m6Am dysregulation in brain, and may be involved in the pathogenesis of depression. The study of RNA methylations in depression has further deepened our understanding of the pathogenesis and development process of depression, provides new perspectives for the study of the pathological mechanism of depression, and provides new targets for the prevention and treatment of this disease.
Collapse
Affiliation(s)
- Chen Li
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Pengfei Xu
- Department of Orthopedics, the First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
| | - Yurong Huang
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Yuting Wang
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Yajie Wu
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Hui Li
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Yanhui Peng
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Xiao Wang
- Department of Clinical Nursing, Anhui University of Chinese Medicine, Hefei, Anhui, China.
| | - Guoying Li
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China.
| | - Juan He
- Department of Gynecology, Anhui Maternal and Child Health Hospital, Hefei, Anhui, China.
| | - Chenggui Miao
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China; Institute of Traditional Chinese Medicine for the Prevention and Treatment of Rheumatology, Anhui University of Chinese Medicine, Hefei, Anhui, China.
| |
Collapse
|
6
|
Guerra-Cantera S, Frago LM, Jiménez-Hernaiz M, Collado-Pérez R, Canelles S, Ros P, García-Piqueras J, Pérez-Nadador I, Barrios V, Argente J, Chowen JA. The metabolic effects of resumption of a high fat diet after weight loss are sex dependent in mice. Sci Rep 2023; 13:13227. [PMID: 37580448 PMCID: PMC10425431 DOI: 10.1038/s41598-023-40514-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 08/11/2023] [Indexed: 08/16/2023] Open
Abstract
Dietary restriction is a frequent strategy for weight loss, but adherence is difficult and returning to poor dietary habits can result in more weight gain than that previously lost. How weight loss due to unrestricted intake of a healthy diet affects the response to resumption of poor dietary habits is less studied. Moreover, whether this response differs between the sexes and if the insulin-like growth factor (IGF) system, sex dependent and involved in metabolic control, participates is unknown. Mice received rodent chow (6% Kcal from fat) or a high-fat diet (HFD, 62% Kcal from fat) for 4 months, chow for 3 months plus 1 month of HFD, or HFD for 2 months, chow for 1 month then HFD for 1 month. Males and females gained weight on HFD and lost weight when returned to chow at different rates (p < 0.001), but weight gain after resumption of HFD intake was not affected by previous weight loss in either sex. Glucose metabolism was more affected by HFD, as well as the re-exposure to HFD after weight loss, in males. This was associated with increases in hypothalamic mRNA levels of IGF2 (p < 0.01) and IGF binding protein (IGFBP) 2 (p < 0.05), factors involved in glucose metabolism, again only in males. Likewise, IGF2 increased IGFBP2 mRNA levels only in hypothalamic astrocytes from males (p < 0.05). In conclusion, the metabolic responses to dietary changes were less severe and more delayed in females and the IGF system might be involved in some of the sex specific observations.
Collapse
Affiliation(s)
- Santiago Guerra-Cantera
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura M Frago
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - María Jiménez-Hernaiz
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Roberto Collado-Pérez
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
| | - Sandra Canelles
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Purificación Ros
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Department of Endocrinology, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Jorge García-Piqueras
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
| | - Iris Pérez-Nadador
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
| | - Vicente Barrios
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain.
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
- IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain.
| | - Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
- IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain.
| |
Collapse
|
7
|
Pahl MC, Grant SFA, Leibel RL, Stratigopoulos G. Technologies, strategies, and cautions when deconvoluting genome-wide association signals: FTO in focus. Obes Rev 2023; 24:e13558. [PMID: 36882962 DOI: 10.1111/obr.13558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 10/08/2022] [Accepted: 01/31/2023] [Indexed: 03/09/2023]
Abstract
Genome-wide association studies have revealed a plethora of genetic variants that correlate with polygenic conditions. However, causal molecular mechanisms have proven challenging to fully define. Without such information, the associations are not physiologically useful or clinically actionable. By reviewing studies of the FTO locus in the genetic etiology of obesity, we wish to highlight advances in the field fueled by the evolution of technical and analytic strategies in assessing the molecular bases for genetic associations. Particular attention is drawn to extrapolating experimental findings from animal models and cell types to humans, as well as technical aspects used to identify long-range DNA interactions and their biological relevance with regard to the associated trait. A unifying model is proposed by which independent obesogenic pathways regulated by multiple FTO variants and genes are integrated at the primary cilium, a cellular antenna where signaling molecules that control energy balance convene.
Collapse
Affiliation(s)
- Matthew C Pahl
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Struan F A Grant
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Division of Diabetes and Endocrinology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Pediatrics, The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA.,Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rudolph L Leibel
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York, USA.,Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, New York, USA
| | - George Stratigopoulos
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York, USA.,Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, New York, USA
| |
Collapse
|
8
|
Moazeny M, Dehbashi M, Hojati Z, Esmaeili F. Investigating neural differentiation of mouse P19 embryonic stem cells in a time-dependent manner by bioinformatic, microscopic and transcriptional analyses. Mol Biol Rep 2023; 50:2183-2194. [PMID: 36565416 DOI: 10.1007/s11033-022-08166-7] [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: 08/16/2021] [Accepted: 09/23/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND As an available cell line, mouse pluripotent P19 has been widely employed for neuronal differentiation studies. In this research, by applying the in vitro differentiation of this cell line into neuron-like cells through retinoic acid (RA) treatment, the roles of some genes including DNMT3B, ICAM1, IRX3, JAK2, LHX1, SOX9, TBX3 and THY1 in neural differentiation was investigated. METHODS AND RESULTS Bioinformatics, microscopic, and transcriptional studies were conducted in a time-dependent manner after RA-induced neural differentiation. According to bioinformatics studies, we determined the engagement of the metabolic and developmental super-pathways and pathways in neural cell differentiation, particularly focusing on the considered genes. According to our qRT-PCR analyses, JAK2, SOX9, TBX3, LHX1 and IRX3 genes were found to be significantly overexpressed in a time-dependent manner (p < 0.05). In addition, the significant downregulation of THY1, DNMT3B and ICAM1 genes was observed during the experiment (p < 0.05). The optical microscopic investigation showed that the specialized extensions of the neuron-like cells were revealed on day 8 after RA treatment. CONCLUSION Accordingly, the neural differentiation of P19 cell line and the role of the considered genes during the differentiation were proved. However, our results warrant further in vivo studies.
Collapse
Affiliation(s)
- Marzieh Moazeny
- Division of Genetics, Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, 81746-73441, Isfahan, Iran
| | - Moein Dehbashi
- Division of Genetics, Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, 81746-73441, Isfahan, Iran
| | - Zohreh Hojati
- Division of Genetics, Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, 81746-73441, Isfahan, Iran.
| | - Fariba Esmaeili
- Division of Animal Sciences, Department of Plant and Animal Biology, Faculty of Biological Science and Technology, University of Isfahan, 81746-73441, Isfahan, Iran
| |
Collapse
|
9
|
The orphan G protein-coupled receptor, GPR139, is expressed in the hypothalamus and is involved in the regulation of body mass, blood glucose, and insulin. Neurosci Lett 2023; 792:136955. [PMID: 36347339 DOI: 10.1016/j.neulet.2022.136955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/19/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022]
Abstract
GPR139 is an orphan G-protein-coupled receptor that is expressed in restricted areas of the nervous system, including the hypothalamus. In this study, we hypothesized that GPR139 could be involved in the regulation of energy balance and metabolism. In the first part of the study, we confirmed that GPR139 is expressed in the hypothalamus and particularly in proopiomelanocortin and agouti-related peptide neurons of the mediobasal hypothalamus. Using a lentivirus with a short-hairpin RNA, we inhibited the expression of GPR139 bilaterally in the mediobasal hypothalamus of mice. The intervention promoted a 40% reduction in the hypothalamic expression of GPR139, which was accompanied by an increase in body mass, a reduction in fasting blood glucose levels, and an increase in insulin levels. In the hypothalamus, inhibition of GPR139 was accompanied by a reduction in the expression of orexin. As previous studies using a pharmacological antagonist of orexin showed a beneficial impact on type 2 diabetes and glucose metabolism, we propose that the inhibition of hypothalamic GPR139 could be acting indirectly through the orexin system to control systemic glucose and insulin. In conclusion, this study advances the characterization of GPR139 in the hypothalamus, demonstrating its involvement in the regulation of body mass, blood insulin, and glycemia.
Collapse
|
10
|
Vinnicombe KRT, Volkoff H. Possible role of transcription factors (BSX, NKX2.1, IRX3 and SIRT1) in the regulation of appetite in goldfish (Carassius auratus). Comp Biochem Physiol A Mol Integr Physiol 2022; 268:111189. [PMID: 35307341 DOI: 10.1016/j.cbpa.2022.111189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/13/2022] [Accepted: 03/13/2022] [Indexed: 12/17/2022]
Abstract
The homeobox genes play important roles in the embryonic development of animals. Recent evidence suggests they might also regulate feeding and act as transcription factors of appetite regulators. Examples of these genes are a brain-specific homeobox transcription factor (BSX), NK2 homeobox 1 (NKX2.1) and the Iroquois homeobox 3 (IRX3). Sirtuin1 (SIRT1) acts as a transcription factor for nutrient (e.g. lipid, glucose) homeostasis and responds to stress and nutrient availability, and has been shown to interact with appetite regulators. Very little is known about the role of these genes in the regulation of feeding and nutrient homeostasis in fish. In this study, we assessed the roles of BSX, NKX2.1, IRX3 and SIRT1 in the central regulation of feeding in goldfish by examining their mRNA brain distribution, assessing the effects of fasting on their brain expression and assessing the effects of peripheral injections of cholecystokinin (CCK, a brain-gut peptide), on their brain expression. All genes showed a widespread distribution in the brain, with high levels in the hypothalamus. In both hypothalamus and telencephalon, fasting induced increases in BSX, IRX3 and NKX2.1 expressions but had no effect on SIRT1 expression levels. CCK injections increased hypothalamic expression levels of IRX3 and SIRT1, and telencephalic expression levels of NKX2.1 and SIRT1, with no effect on either hypothalamic BSX or NKX2.1 expression levels or telencephalon BSX or IRX3 expression levels. Our results suggest that, in goldfish as in mammals, central BSX, NKX2.1, IRX3 and SIRT1 are present in regions of the brain regulating feeding, are sensitive to nutrient status and interact with appetite-regulating peptides.
Collapse
Affiliation(s)
- Kelsey R T Vinnicombe
- Department of Biology, Memorial University of Newfoundland, St. John's, NL A1B 3X9, Canada
| | - Helene Volkoff
- Department of Biology, Memorial University of Newfoundland, St. John's, NL A1B 3X9, Canada.
| |
Collapse
|
11
|
Yang Z, Yu GL, Zhu X, Peng TH, Lv YC. Critical roles of FTO-mediated mRNA m6A demethylation in regulating adipogenesis and lipid metabolism: Implications in lipid metabolic disorders. Genes Dis 2022; 9:51-61. [PMID: 35005107 PMCID: PMC8720706 DOI: 10.1016/j.gendis.2021.01.005] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/19/2020] [Accepted: 01/20/2021] [Indexed: 02/06/2023] Open
Abstract
The goal this review is to clarify the effects of the fat mass and obesity-associated protein (FTO) in lipid metabolism regulation and related underlying mechanisms through the FTO-mediated demethylation of m6A modification. FTO catalyzes the demethylation of m6A to alter the processing, maturation and translation of the mRNAs of lipid-related genes. FTO overexpression in the liver promotes lipogenesis and lipid droplet (LD) enlargement and suppresses CPT-1–mediated fatty acid oxidation via the SREBP1c pathway, promoting excessive lipid storage and nonalcoholic fatty liver diseases (NAFLD). FTO enhances preadipocyte differentiation through the C/EBPβ pathway, and facilitates adipogenesis and fat deposition by altering the alternative splicing of RUNX1T1, the expression of PPARγ and ANGPTL4, and the phosphorylation of PLIN1, whereas it inhibits lipolysis by inhibiting IRX3 expression and the leptin pathway, causing the occurrence and development of obesity. Suppression of the PPARβ/δ and AMPK pathways by FTO-mediated m6A demethylation damages lipid utilization in skeletal muscles, leading to the occurrence of diabetic hyperlipidemia. m6A demethylation by FTO inhibits macrophage lipid influx by downregulating PPARγ protein expression and accelerates cholesterol efflux by phosphorylating AMPK, thereby impeding foam cell formation and atherosclerosis development. In summary, FTO-mediated m6A demethylation modulates the expression of lipid-related genes to regulate lipid metabolism and lipid disorder diseases.
Collapse
Affiliation(s)
- Zhou Yang
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Guang-Li Yu
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Xiao Zhu
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi 541199, PR China
| | - Tian-Hong Peng
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Yun-Cheng Lv
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China.,Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi 541199, PR China
| |
Collapse
|
12
|
Ma Y, Chen G, Yi J, Li Q, Tan Z, Fan W, Luo X, He Z, Si Z, Li J. IRX3 plays an important role in the pathogenesis of metabolic-associated fatty liver disease by regulating hepatic lipid metabolism. Front Endocrinol (Lausanne) 2022; 13:895593. [PMID: 35957832 PMCID: PMC9360787 DOI: 10.3389/fendo.2022.895593] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 07/01/2022] [Indexed: 12/13/2022] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD) affects approximately a quarter of the global population. Identification of the key genes and pathways involved in hepatic lipid metabolism is of the utmost importance for the diagnosis, treatment, and prevention of MAFLD. In this study, differentially expressed genes were identified through whole-genome transcriptional analysis of liver tissue from MAFLD patients and healthy controls, and a series of lipid metabolism-related molecules and pathways were obtained through pathway analysis. Subsequently, we focused on Iroquois homeobox protein 3 (IRX3), one of 13 transcription factors that were screened from the 331 differentially expressed genes. The transcription factor IRX3 was significantly decreased in the liver tissue of patients with MAFLD when compared with healthy controls. Pearson's correlation analysis showed that the expression levels of IRX3 in liver tissue were negatively correlated with serum total cholesterol, triglycerides, low-density lipoprotein cholesterol, and uric acid levels. The overexpression and interference of IRX3 induced the increased and decreased lipid droplet accumulation in vitro, respectively. Moreover, interference of IRX3 expression increased mitochondrial fragmentation and reduced the activity of the mitochondrial respiratory chain complex IV. In summary, the study demonstrated that IRX3 regulated hepatic lipid metabolism of MAFLD, and also revealed the effect of IRX3 on mitochondria might be an important mechanism by which IRX3 regulated hepatic lipid metabolism of MAFLD.
Collapse
Affiliation(s)
- Yongqiang Ma
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, Changsha, China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Guangshun Chen
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, Changsha, China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, China
- Transplant Medical Research Center, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Junfang Yi
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Qiang Li
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, Changsha, China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, China
- Transplant Medical Research Center, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhi Tan
- Department of Gastroenterology, The First Hospital of Changsha, Changsha, China
| | - Wenling Fan
- Department of Gastroenterology, The First Hospital of Changsha, Changsha, China
| | - Xiaohua Luo
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhiyong He
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhongzhou Si
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, Changsha, China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, China
- Transplant Medical Research Center, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jiequn Li
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, Changsha, China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, China
- Transplant Medical Research Center, The Second Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Jiequn Li,
| |
Collapse
|
13
|
Dou Z, Son JE, Hui CC. Irx3 and Irx5 - Novel Regulatory Factors of Postnatal Hypothalamic Neurogenesis. Front Neurosci 2021; 15:763856. [PMID: 34795556 PMCID: PMC8593166 DOI: 10.3389/fnins.2021.763856] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/07/2021] [Indexed: 12/27/2022] Open
Abstract
The hypothalamus is a brain region that exhibits highly conserved anatomy across vertebrate species and functions as a central regulatory hub for many physiological processes such as energy homeostasis and circadian rhythm. Neurons in the arcuate nucleus of the hypothalamus are largely responsible for sensing of peripheral signals such as leptin and insulin, and are critical for the regulation of food intake and energy expenditure. While these neurons are mainly born during embryogenesis, accumulating evidence have demonstrated that neurogenesis also occurs in postnatal-adult mouse hypothalamus, particularly in the first two postnatal weeks. This second wave of active neurogenesis contributes to the remodeling of hypothalamic neuronal populations and regulation of energy homeostasis including hypothalamic leptin sensing. Radial glia cell types, such as tanycytes, are known to act as neuronal progenitors in the postnatal mouse hypothalamus. Our recent study unveiled a previously unreported radial glia-like neural stem cell (RGL-NSC) population that actively contributes to neurogenesis in the postnatal mouse hypothalamus. We also identified Irx3 and Irx5, which encode Iroquois homeodomain-containing transcription factors, as genetic determinants regulating the neurogenic property of these RGL-NSCs. These findings are significant as IRX3 and IRX5 have been implicated in FTO-associated obesity in humans, illustrating the importance of postnatal hypothalamic neurogenesis in energy homeostasis and obesity. In this review, we summarize current knowledge regarding postnatal-adult hypothalamic neurogenesis and highlight recent findings on the radial glia-like cells that contribute to the remodeling of postnatal mouse hypothalamus. We will discuss characteristics of the RGL-NSCs and potential actions of Irx3 and Irx5 in the regulation of neural stem cells in the postnatal-adult mouse brain. Understanding the behavior and regulation of neural stem cells in the postnatal-adult hypothalamus will provide novel mechanistic insights in the control of hypothalamic remodeling and energy homeostasis.
Collapse
Affiliation(s)
- Zhengchao Dou
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Joe Eun Son
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Chi-chung Hui
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
14
|
Yao J, Wu D, Zhang C, Yan T, Zhao Y, Shen H, Xue K, Huang X, Wang Z, Qiu Y. Macrophage IRX3 promotes diet-induced obesity and metabolic inflammation. Nat Immunol 2021; 22:1268-1279. [PMID: 34556885 DOI: 10.1038/s41590-021-01023-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 08/04/2021] [Indexed: 02/07/2023]
Abstract
Metabolic inflammation is closely linked to obesity, and is implicated in the pathogenesis of metabolic diseases. FTO harbors the strongest genetic association with polygenic obesity, and IRX3 mediates the effects of FTO on body weight. However, in what cells and how IRX3 carries out this control are poorly understood. Here we report that macrophage IRX3 promotes metabolic inflammation to accelerate the development of obesity and type 2 diabetes. Mice with myeloid-specific deletion of Irx3 were protected against diet-induced obesity and metabolic diseases via increasing adaptive thermogenesis. Mechanistically, macrophage IRX3 promoted proinflammatory cytokine transcription and thus repressed adipocyte adrenergic signaling, thereby inhibiting lipolysis and thermogenesis. JNK1/2 phosphorylated IRX3, leading to its dimerization and nuclear translocation for transcription. Further, lipopolysaccharide stimulation stabilized IRX3 by inhibiting its ubiquitination, which amplified the transcriptional capacity of IRX3. Together, our findings identify a new player, macrophage IRX3, in the control of body weight and metabolic inflammation, implicating IRX3 as a therapeutic target.
Collapse
Affiliation(s)
- Jingfei Yao
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Dongmei Wu
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Chunyan Zhang
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Ting Yan
- School of life Sciences, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Yiheng Zhao
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.,Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Hongyu Shen
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.,Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Kaili Xue
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Xun Huang
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.,Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Zihao Wang
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Yifu Qiu
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing, China. .,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.
| |
Collapse
|
15
|
The IRX(3)some factor in macrophages. Nat Immunol 2021; 22:1203-1204. [PMID: 34556882 DOI: 10.1038/s41590-021-01032-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
16
|
Son JE, Dou Z, Kim KH, Wanggou S, Cha VSB, Mo R, Zhang X, Chen X, Ketela T, Li X, Huang X, Hui CC. Irx3 and Irx5 in Ins2-Cre + cells regulate hypothalamic postnatal neurogenesis and leptin response. Nat Metab 2021; 3:701-713. [PMID: 33859429 DOI: 10.1038/s42255-021-00382-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 03/08/2021] [Indexed: 12/24/2022]
Abstract
Obesity is mainly due to excessive food intake. IRX3 and IRX5 have been suggested as determinants of obesity in connection with the intronic variants of FTO, but how these genes contribute to obesity via changes in food intake remains unclear. Here, we show that mice doubly heterozygous for Irx3 and Irx5 mutations exhibit lower food intake with enhanced hypothalamic leptin response. By lineage tracing and single-cell RNA sequencing using the Ins2-Cre system, we identify a previously unreported radial glia-like neural stem cell population with high Irx3 and Irx5 expression in early postnatal hypothalamus and demonstrate that reduced dosage of Irx3 and Irx5 promotes neurogenesis in postnatal hypothalamus leading to elevated numbers of leptin-sensing arcuate neurons. Furthermore, we find that mice with deletion of Irx3 in these cells also exhibit a similar food intake and hypothalamic phenotype. Our results illustrate that Irx3 and Irx5 play a regulatory role in hypothalamic postnatal neurogenesis and leptin response.
Collapse
Affiliation(s)
- Joe Eun Son
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Zhengchao Dou
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Kyoung-Han Kim
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
- University of Ottawa Heart Institute and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Siyi Wanggou
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Vincent Su Bin Cha
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Rong Mo
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Xiaoyun Zhang
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Xinyu Chen
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Troy Ketela
- Princess Margaret Genomics Centre, University Health Network, Toronto, Ontario, Canada
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xi Huang
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Chi-Chung Hui
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
17
|
Variable expression quantitative trait loci analysis of breast cancer risk variants. Sci Rep 2021; 11:7192. [PMID: 33785833 PMCID: PMC8009949 DOI: 10.1038/s41598-021-86690-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 03/12/2021] [Indexed: 01/08/2023] Open
Abstract
Genome wide association studies (GWAS) have identified more than 180 variants associated with breast cancer risk, however the underlying functional mechanisms and biological pathways which confer disease susceptibility remain largely unknown. As gene expression traits are under genetic regulation we hypothesise that differences in gene expression variability may identify causal breast cancer susceptibility genes. We performed variable expression quantitative trait loci (veQTL) analysis using tissue-specific expression data from the Genotype-Tissue Expression (GTEx) Common Fund Project. veQTL analysis identified 70 associations (p < 5 × 10–8) consisting of 60 genes and 27 breast cancer risk variants, including 55 veQTL that were observed in breast tissue only. Pathway analysis of genes associated with breast-specific veQTL revealed an enrichment of four genes (CYP11B1, CYP17A1 HSD3B2 and STAR) involved in the C21-steroidal biosynthesis pathway that converts cholesterol to breast-related hormones (e.g. oestrogen). Each of these four genes were significantly more variable in individuals homozygous for rs11075995 (A/A) breast cancer risk allele located in the FTO gene, which encodes an RNA demethylase. The A/A allele was also found associated with reduced expression of FTO, suggesting an epi-transcriptomic mechanism may underlie the dysregulation of genes involved in hormonal biosynthesis leading to an increased risk of breast cancer. These findings provide evidence that genetic variants govern high levels of expression variance in breast tissue, thus building a more comprehensive insight into the underlying biology of breast cancer risk loci.
Collapse
|
18
|
Monfort-Pires M, U-Din M, Nogueira GA, de Almeida-Faria J, Sidarta-Oliveira D, Sant'Ana MR, De Lima-Júnior JC, Cintra DE, de Souza HP, Ferreira SRG, Sapienza MT, Virtanen KA, Velloso LA. Short Dietary Intervention with Olive Oil Increases Brown Adipose Tissue Activity in Lean but not Overweight Subjects. J Clin Endocrinol Metab 2021; 106:472-484. [PMID: 33180910 DOI: 10.1210/clinem/dgaa824] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Indexed: 01/09/2023]
Abstract
BACKGROUND The brown adipose tissue (BAT) is a potential target for the treatment of obesity and metabolic disorders. Its activation by cold exposure or adrenergic drugs can increase systemic insulin sensitivity and improve lipid metabolism; however, little is known about the effects of specific dietary components on BAT activity. OBJECTIVES We asked if a short-term (4 weeks) dietary intervention with olive oil could modify BAT activity in lean and overweight/obese volunteers. DESIGN This was a 4-week open clinical trial in which all participants underwent a dietary intervention with extra-virgin olive oil supplementation. As the initial intake of olive oil was controlled all the participants were controls of themselves. RESULTS The intervention resulted in significant increase in blood monounsaturated fatty acid levels, which was accompanied by increased BAT activity in lean but not in overweight/obese volunteers. In the lean group, an increase in leptin was detected after the intervention, and low leptin values at the beginning of the study were predictive of greater BAT activity after intervention. In addition, increase in leptin concentration was associated with increased BAT activity. Three known endogenous mediators of BAT activity, secretin, fibroblast growth factor 21 (FGF21), and 12,13-dihydroxy-9Z-octadecenoic acid (12,13-diHOME) were increased by intervention in lean, whereas only secretin and FGF21 were increased in subjects with excessive weight. CONCLUSION This study provides clinical evidence for the impact of monounsaturated fatty acids on BAT activity and an advance in the understanding of the beneficial health effects of olive oil.
Collapse
Affiliation(s)
- Milena Monfort-Pires
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Mueez U-Din
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Guilherme A Nogueira
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Juliana de Almeida-Faria
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Davi Sidarta-Oliveira
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Marcella Ramos Sant'Ana
- Nutritional Genomics Laboratory, LabGeN, School of Applied Sciences, UNICAMP, Limeira, São Paulo, Brazil
| | - José C De Lima-Júnior
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Dennys E Cintra
- Nutritional Genomics Laboratory, LabGeN, School of Applied Sciences, UNICAMP, Limeira, São Paulo, Brazil
| | | | - Sandra R G Ferreira
- Department of Epidemiology, School of Public Health - University of São Paulo, São Paulo, SP, Brazil
| | - Marcelo Tatit Sapienza
- Division of Nuclear Medicine, Department of Radiology and Oncology, Medical School of University of São Paulo (FMUSP), São Paulo, Brazil
| | - Kirsi A Virtanen
- Turku PET Centre, Turku University Hospital, Turku, Finland
- Clinical Nutrition, Institute of Public Health and Clinical Nutrition, University of Eastern Finland (UEF), Kuopio, Finland
| | - Licio A Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| |
Collapse
|
19
|
Zhang Z, Wu Q, He Y, Lu P, Li D, Yang M, Gu W, Liu R, Hong J, Wang J. IRX3 Overexpression Enhances Ucp1 Expression In Vivo. Front Endocrinol (Lausanne) 2021; 12:634191. [PMID: 33776928 PMCID: PMC7988233 DOI: 10.3389/fendo.2021.634191] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 02/04/2021] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE The Iroquois homeobox 3 (IRX3) gene was recently reported to be a functional downstream target of a common polymorphism in the FTO gene, which encodes an obesity-associated protein; however, the role of IRX3 in energy expenditure remains unclear. Studies have revealed that the overexpression of a dominant-negative form of IRX3 in the mouse hypothalamus and adipose tissue promoted energy expenditure by enhancing brown/browning activities. Meanwhile, we and others recently demonstrated that IRX3 knockdown impaired the browning program of primary preadipocytes in vitro. In this study, we aimed to further clarify the effects of overexpressing human IRX3 (hIRX3) on brown/beige adipose tissues in vivo. METHODS Brown/beige adipocyte-specific hIRX3-overexpressing mice were generated and the browning program of white adipose tissues was induced by both chronic cold stimulation and CL316,243 injection. Body weight, fat mass, lean mass, and energy expenditure were measured, while morphological changes and the expression of thermogenesis-related genes in adipose tissue were analyzed. Moreover, the browning capacity of primary preadipocytes derived from hIRX3-overexpressing mice was assessed. RNA sequencing was also employed to investigate the effect of hIRX3 on the expression of thermogenesis-related genes. RESULTS hIRX3 overexpression in embryonic brown/beige adipose tissues (Rosa26hIRX3 ;Ucp1-Cre) led to increased energy expenditure, decreased fat mass, and a lean body phenotype. After acute cold exposure or CL316,243 stimulation, brown/beige tissue hIRX3-overexpressing mice showed an increase in Ucp1 expression. Consistent with this, induced hIRX3 overexpression in adult mice (Rosa26hIRX3 ;Ucp1-CreERT2) also promoted a moderate increase in Ucp1 expression. Ex vitro experiments further revealed that hIRX3 overexpression induced by Ucp1-driven Cre recombinase activity upregulated brown/beige adipocytes Ucp1 expression and oxygen consumption rate (OCR). RNA sequencing analyses indicated that hIRX3 overexpression in brown adipocytes enhanced brown fat cell differentiation, glycolysis, and gluconeogenesis. CONCLUSION Consistent with the in vitro findings, brown/beige adipocyte-specific overexpression of hIRX3 promoted Ucp1 expression and thermogenesis, while reducing fat mass.
Collapse
Affiliation(s)
- Zhiyin Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qihan Wu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang He
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peng Lu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Danjie Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Minglan Yang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqiong Gu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruixin Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Hong
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiqiu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
20
|
Chaves FM, Mansano NS, Frazão R, Donato J. Tumor Necrosis Factor α and Interleukin-1β Acutely Inhibit AgRP Neurons in the Arcuate Nucleus of the Hypothalamus. Int J Mol Sci 2020; 21:ijms21238928. [PMID: 33255553 PMCID: PMC7728092 DOI: 10.3390/ijms21238928] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/31/2022] Open
Abstract
Obesity-associated low-grade inflammation favors weight gain, whereas systemic infection frequently leads to anorexia. Thus, inflammatory signals can either induce positive or negative energy balance. In this study, we used whole-cell patch-clamp to investigate the acute effects of three important proinflammatory cytokines, tumor necrosis factor α (TNF-α), interleukin-6, and interleukin-1β (IL-1β) on the membrane excitability of agouti-related peptide (AgRP)- or proopiomelanocortin (POMC)-producing neurons. We found that both TNF-α and IL-1β acutely inhibited the activity of 35-42% of AgRP-producing neurons, whereas very few POMC neurons were depolarized by TNF-α. Interleukin-6 induced no acute changes in the activity of AgRP or POMC neurons. Our findings indicate that the effect of TNF-α and IL-1β, especially on the activity of AgRP-producing neurons, may contribute to inflammation-induced anorexia observed during acute inflammatory conditions.
Collapse
Affiliation(s)
- Fernanda M. Chaves
- Departamento de Fisiologia e Biofísica, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil;
| | - Naira S. Mansano
- Departamento de Anatomia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-900, Brazil;
| | - Renata Frazão
- Departamento de Anatomia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-900, Brazil;
- Correspondence: (R.F.); (J.D.J.)
| | - Jose Donato
- Departamento de Fisiologia e Biofísica, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil;
- Correspondence: (R.F.); (J.D.J.)
| |
Collapse
|
21
|
Guerra-Cantera S, Frago LM, Jiménez-Hernaiz M, Ros P, Freire-Regatillo A, Barrios V, Argente J, Chowen JA. Impact of Long-Term HFD Intake on the Peripheral and Central IGF System in Male and Female Mice. Metabolites 2020; 10:metabo10110462. [PMID: 33202914 PMCID: PMC7698111 DOI: 10.3390/metabo10110462] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022] Open
Abstract
The insulin-like growth factor (IGF) system is responsible for growth, but also affects metabolism and brain function throughout life. New IGF family members (i.e., pappalysins and stanniocalcins) control the availability/activity of IGFs and are implicated in growth. However, how diet and obesity modify this system has been poorly studied. We explored how intake of a high-fat diet (HFD) or commercial control diet (CCD) affects the IGF system in the circulation, visceral adipose tissue (VAT) and hypothalamus. Male and female C57/BL6J mice received HFD (60% fat, 5.1 kcal/g), CCD (10% fat, 3.7 kcal/g) or chow (3.1 % fat, 3.4 kcal/g) for 8 weeks. After 7 weeks of HFD intake, males had decreased glucose tolerance (p < 0.01) and at sacrifice increased plasma insulin (p < 0.05) and leptin (p < 0.01). Circulating free IGF1 (p < 0.001), total IGF1 (p < 0.001), IGF2 (p < 0.05) and IGFBP3 (p < 0.01) were higher after HFD in both sexes, with CCD increasing IGFBP2 in males (p < 0.001). In VAT, HFD reduced mRNA levels of IGF2 (p < 0.05), PAPP-A (p < 0.001) and stanniocalcin (STC)-1 (p < 0.001) in males. HFD increased hypothalamic IGF1 (p < 0.01), IGF2 (p < 0.05) and IGFBP5 (p < 0.01) mRNA levels, with these changes more apparent in females. Our results show that diet-induced changes in the IGF system are tissue-, sex- and diet-dependent.
Collapse
Affiliation(s)
- Santiago Guerra-Cantera
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009 Madrid, Spain; (S.G.-C.); (L.M.F.); (M.J.-H.); (A.F.-R.); (V.B.)
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, E-28029 Madrid, Spain;
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Laura M. Frago
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009 Madrid, Spain; (S.G.-C.); (L.M.F.); (M.J.-H.); (A.F.-R.); (V.B.)
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, E-28029 Madrid, Spain;
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - María Jiménez-Hernaiz
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009 Madrid, Spain; (S.G.-C.); (L.M.F.); (M.J.-H.); (A.F.-R.); (V.B.)
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Purificación Ros
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, E-28029 Madrid, Spain;
- Department of Pediatrics, Hospital Universitario Puerta de Hierro-Majadahonda, E-28222 Madrid, Spain
| | - Alejandra Freire-Regatillo
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009 Madrid, Spain; (S.G.-C.); (L.M.F.); (M.J.-H.); (A.F.-R.); (V.B.)
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, E-28029 Madrid, Spain;
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Vicente Barrios
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009 Madrid, Spain; (S.G.-C.); (L.M.F.); (M.J.-H.); (A.F.-R.); (V.B.)
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009 Madrid, Spain; (S.G.-C.); (L.M.F.); (M.J.-H.); (A.F.-R.); (V.B.)
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, E-28029 Madrid, Spain;
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
- IMDEA Food Institute, CEI UAM + CSIC, Carretera de Cantoblanco 8, E-28049 Madrid, Spain
- Correspondence: (J.A.); (J.A.C.)
| | - Julie A. Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009 Madrid, Spain; (S.G.-C.); (L.M.F.); (M.J.-H.); (A.F.-R.); (V.B.)
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
- IMDEA Food Institute, CEI UAM + CSIC, Carretera de Cantoblanco 8, E-28049 Madrid, Spain
- Correspondence: (J.A.); (J.A.C.)
| |
Collapse
|
22
|
Reginato A, Siqueira BP, Miyamoto JÉ, Portovedo M, Costa SDO, de Fante T, Rodrigues HG, Ignácio-Souza LM, Torsoni MA, Torsoni AS, Le Stunff H, Belsham DD, Milanski M. Acute effects of fatty acids on autophagy in NPY neurones. J Neuroendocrinol 2020; 32:e12900. [PMID: 33040385 DOI: 10.1111/jne.12900] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 08/15/2020] [Accepted: 08/19/2020] [Indexed: 12/13/2022]
Abstract
High-fat diet (HFD) feeding is deleterious to hypothalamic tissue, leading to inflammation and lipotoxicity, as well as contributing to central insulin resistance. Autophagy is a process that restores cellular homeostasis by degrading malfunctioning organelles and proteins. Chronic HFD-feeding down-regulates hypothalamic autophagy. However, the effects of short-term HFD-feeding and the saturated fatty acid palmitate (PA) on hypothalamic autophagy and in neurones that express neuropeptide Y (NPY) and agouti-related peptide remains unknown. Therefore, we assessed hypothalamic autophagy after 1 and 3 days of HFD-feeding. We also injected PA i.c.v and analysed the modulation of autophagy in hypothalamic tissue. Both interventions resulted in changes in autophagy-related gene profiles without significant differences in protein content of p62 and LC3B-II, markers of the autophagy pathway. When we assessed native NPY neurones in brain slices from PA-treated animals, we observed increased levels of Atg7 and LC3B protein in response to PA treatment, indicating the induction of autophagy. We then tested the direct effects of fatty acids using the immortalised hypothalamic NPY-expressing neuronal cell model mHypoE-46. We found that PA, but not palmitoleate (PO) (a monounsaturated fatty acid), was able to induce autophagy. Co-treatment with PA and PO was able to block the PA-mediated induction of autophagy, as assessed by flow cytometry. When the de novo ceramide synthesis pathway was blocked with myriocin pre-treatment, we observed a decrease in PA-mediated induction of autophagy, although there was no change with the toll-like receptor 4 inhibitor, TAK-242. Taken together, these findings provide evidence that saturated and unsaturated fatty acids can differentially regulate hypothalamic autophagy and that ceramide synthesis may be an important mediator of those effects. Understanding the mechanisms by which dietary fats affect autophagy in neurones involved in the control of energy homeostasis will provide potential new pathways for targeting and containing the obesity epidemic.
Collapse
Affiliation(s)
- Andressa Reginato
- Faculty of Applied Science, University of Campinas, Campinas, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Beatriz Piatezzi Siqueira
- Faculty of Applied Science, University of Campinas, Campinas, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Josiane Érica Miyamoto
- Faculty of Applied Science, University of Campinas, Campinas, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Mariana Portovedo
- Faculty of Applied Science, University of Campinas, Campinas, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Suleyma de Oliveira Costa
- Faculty of Applied Science, University of Campinas, Campinas, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Thaís de Fante
- Faculty of Applied Science, University of Campinas, Campinas, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | | | - Letícia Martins Ignácio-Souza
- Faculty of Applied Science, University of Campinas, Campinas, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Márcio Alberto Torsoni
- Faculty of Applied Science, University of Campinas, Campinas, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Adriana Souza Torsoni
- Faculty of Applied Science, University of Campinas, Campinas, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Hervé Le Stunff
- Neuroscience Institute, Université Paris Saclay, Paris, France
| | - Denise D Belsham
- Departments of Physiology, Ob/Gyn, and Medicine, University of Toronto, Toronto, ON, Canada
| | - Marciane Milanski
- Faculty of Applied Science, University of Campinas, Campinas, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| |
Collapse
|
23
|
Chen EY, Zeffiro TA. Hunger and BMI modulate neural responses to sweet stimuli: fMRI meta-analysis. Int J Obes (Lond) 2020; 44:1636-1652. [PMID: 32555497 PMCID: PMC8023765 DOI: 10.1038/s41366-020-0608-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 02/16/2020] [Accepted: 02/21/2020] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Consuming sweet foods, even when sated, can lead to unwanted weight gain. Contextual factors, such as longer time fasting, subjective hunger, and body mass index (BMI), may increase the likelihood of overeating. Nevertheless, the neural mechanisms underlying these moderating influences on energy intake are poorly understood. METHODS We conducted both categorical meta-analysis and meta-regression of factors modulating neural responses to sweet stimuli, using data from 30 functional magnetic resonance imaging (fMRI) articles incorporating 39 experiments (N = 995) carried out between 2006 and 2019. RESULTS Responses to sweet stimuli were associated with increased activity in regions associated with taste, sensory integration, and reward processing. These taste-evoked responses were modulated by context. Longer fasts were associated with higher posterior cerebellar, thalamic, and striatal activity. Greater self-reported hunger was associated with higher medial orbitofrontal cortex (OFC), dorsal striatum, and amygdala activity and lower posterior cerebellar activity. Higher BMI was associated with higher posterior cerebellar and insular activity. CONCLUSIONS Variations in fasting time, self-reported hunger, and BMI are contexts associated with differential sweet stimulus responses in regions associated with reward processing and homeostatic regulation. These results are broadly consistent with a hierarchical model of taste processing. Hunger, but not fasting or BMI, was associated with sweet stimulus-related OFC activity. Our findings extend existing models of taste processing to include posterior cerebellar regions that are associated with moderating effects of both state (fast length and self-reported hunger) and trait (BMI) variables.
Collapse
Affiliation(s)
- Eunice Y Chen
- TEDP (Temple Eating Disorders Program), Department of Psychology, Temple University, 1701 N 13th Street, Philadelphia, PA, 19122, USA.
| | | |
Collapse
|
24
|
Mariano IR, Yamada LA, Soares Rabassi R, Rissi Sabino VL, Bataglini C, Azevedo SCSF, Garcia RF, Pedrosa MMD. Differential Responses of Liver and Hypothalamus to the Nutritional Condition During Lactation and Adult Life. Front Physiol 2020; 11:553. [PMID: 32581843 PMCID: PMC7291834 DOI: 10.3389/fphys.2020.00553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 04/30/2020] [Indexed: 01/21/2023] Open
Abstract
It was previously reported that liver glucose metabolism in rats under caloric restriction differs from that of freely-fed rats. This study hypothesized that these changes (1) were related to the expression of hypothalamic neuropeptides involved in metabolic control, and (2) were not a residual effect of litter size. To those purposes, liver glucose metabolism and hypothalamic expression of the orexigenic neuropeptides NPY (neuropeptide Y) and AgRP (agouti gene-related peptide); and of the anorexigenic neuropeptides POMC (pro-opiomelanocortin) and CART (cocaine- and amphetamine-related transcripts) were investigated. Male Wistar rats from two different litter sizes (G6 and G12, with 6 or 12 pups, respectively) were subjected to free feeding (GL, ad libitum), 50% caloric restriction (GR) or caloric restriction+ad libitum refeeding (GRL) until the age of 90 days. Biometric values were lower in GR than in GL, while in GRL they were totally or partially recovered. Blood glucose variation during the pyruvate tolerance test (PTT) was small in GR. During in situ liver perfusion, total, basal, and adrenaline-stimulated liver glucose outputs were high in GR, but additional glucose output in the presence of alanine was negligible. Refeeding (GRL) yielded values close to those of GL. Litter size did not consistently influence any of these variables. The expression of transcripts of the hypothalamic neuropeptides was responsive to feeding regimen, litter size and/or their interaction and differed from G6 to G12, while the metabolic changes of the liver were qualitatively equal in both GR. Therefore, the changes in glucose metabolism in the liver of rats under caloric restriction were not determined by either litter size or hypothalamic neuropeptide expression and were linked only to the prevailing feeding regimen of the adult animal.
Collapse
Affiliation(s)
- Isabela Ramos Mariano
- Laboratory of Physiological Sciences and Hepatic Metabolism, Department of Physiological Sciences, State University of Maringá, Maringá, Brazil
| | - Laís Akemi Yamada
- Laboratory of Physiological Sciences and Hepatic Metabolism, Department of Physiological Sciences, State University of Maringá, Maringá, Brazil
| | - Renan Soares Rabassi
- Laboratory of Physiological Sciences and Hepatic Metabolism, Department of Physiological Sciences, State University of Maringá, Maringá, Brazil
| | - Vanessa Lara Rissi Sabino
- Laboratory of Physiological Sciences and Hepatic Metabolism, Department of Physiological Sciences, State University of Maringá, Maringá, Brazil
| | - Camila Bataglini
- Laboratory of Physiological Sciences and Hepatic Metabolism, Department of Physiological Sciences, State University of Maringá, Maringá, Brazil
| | | | - Rosângela Fernandes Garcia
- Laboratory of Physiological Sciences and Hepatic Metabolism, Department of Physiological Sciences, State University of Maringá, Maringá, Brazil
| | - Maria Montserrat Diaz Pedrosa
- Laboratory of Physiological Sciences and Hepatic Metabolism, Department of Physiological Sciences, State University of Maringá, Maringá, Brazil
| |
Collapse
|
25
|
de Araújo TM, Velloso LA. Hypothalamic IRX3: A New Player in the Development of Obesity. Trends Endocrinol Metab 2020; 31:368-377. [PMID: 32035736 DOI: 10.1016/j.tem.2020.01.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 01/06/2020] [Accepted: 01/09/2020] [Indexed: 12/15/2022]
Abstract
Genome-wide association studies (GWASs) have identified SNPs of the fat mass and obesity (FTO) gene as the most important risk alleles for obesity. However, how the presence of risk alleles affect phenotype is still a matter of intense investigation. In 2014, a study revealed that long-range enhancers from the intronic regions of the FTO gene regulate iroquois-class homeobox protein (IRX)3 expression. IRX3 is expressed in hypothalamic pro-opiomelanocortin (POMC) neurons and changes in its expression levels affect body adiposity by modifying food intake and energy expenditure. These findings have placed IRX3 as a potential target for the treatment of obesity. Here, we review studies that evaluated the roles of IRX3 in development, neurogenesis, and body energy homeostasis.
Collapse
Affiliation(s)
- Thiago Matos de Araújo
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Licio A Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas (UNICAMP), Campinas, Brazil.
| |
Collapse
|
26
|
Bjune JI, Dyer L, Røsland GV, Tronstad KJ, Njølstad PR, Sagen JV, Dankel SN, Mellgren G. The homeobox factor Irx3 maintains adipogenic identity. Metabolism 2020; 103:154014. [PMID: 31751577 DOI: 10.1016/j.metabol.2019.154014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 10/22/2019] [Accepted: 11/13/2019] [Indexed: 01/13/2023]
Abstract
BACKGROUND Inhibition of Irx3 and Irx5 has been shown to reduce body weight and white adipose tissue (WAT) mass through cell-autonomous and sympathetic-induced increases in adipocyte beiging and thermogenesis in mice and humans. However, the underlying mechanisms of the Irx control over beiging are still largely unknown, as illustrated by recent reports showing divergent effects of Irx3 on adipocyte metabolism and function. Here, we investigated the role of Irx3 in controlling beige preadipocyte function and differentiation. METHODS Stable knock out of Irx3 in ME3 mouse preadipocytes capable of beiging was performed using a CRISPR-Cas9 system, and the effect on cell differentiation was assessed by qPCR, RNA-seq, Oil-red-O lipid staining and Alcian Blue staining of proteoglycans. Changes in cell identities were validated using cell type enrichment analysis from RNA-seq data. Proliferation and cell cycle progression in undifferentiated cells were measured by WST-1 and flow cytometry, reactive oxygen species (ROS) generation was determined by fluorescence spectrometry and mitochondrial respiration was investigated by Seahorse assay. RESULTS Irx3 was found to be essential for the identity, function and adipogenic differentiation of beige adipocyte precursors. Irx3-KO impaired proliferation, ROS generation and mitochondrial respiration in the preadipocytes. We further observed profound changes in numerous genes during both early and late stages of adipogenic differentiation, including genes important for adipocyte differentiation, cell cycle progression, oxidative phosphorylation (OXPHOS) and morphogenesis. Irx3-KO cells failed to accumulate lipids following adipogenic stimuli, and cell enrichment analysis revealed a loss of preadipocyte identity and a gain of chondrocyte-like identity in Irx3-KO cells during early differentiation. Finally, unlike the control cells, the Irx3-KO cells readily responded to chondrogenic stimuli. CONCLUSIONS Irx3 is required for preadipocyte identity and differentiation capacity. Our findings suggest that, while inhibition of Irx3 may be beneficial during later developmental stages to modulate adipogenesis in the beige direction, constitutive and complete absence of Irx3 in the embryonic fibroblast stage leads to detrimental loss of adipogenic differentiation capacity.
Collapse
Affiliation(s)
- Jan-Inge Bjune
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway; Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway; Hormone Laboratory, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Laurence Dyer
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway; Hormone Laboratory, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Gro V Røsland
- Department of Biomedicine, University of Bergen, N-5020 Bergen, Norway
| | - Karl Johan Tronstad
- Department of Biomedicine, University of Bergen, N-5020 Bergen, Norway; Department of Oncology and Medical Physics, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Pål R Njølstad
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway; Department of Pediatrics and Adolescents, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Jørn V Sagen
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway; Hormone Laboratory, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Simon N Dankel
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway; Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway; Hormone Laboratory, Haukeland University Hospital, N-5021 Bergen, Norway.
| | - Gunnar Mellgren
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway; Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway; Hormone Laboratory, Haukeland University Hospital, N-5021 Bergen, Norway.
| |
Collapse
|
27
|
Meyre D, Mohamed S, Gray JC, Weafer J, MacKillop J, de Wit H. Association between impulsivity traits and body mass index at the observational and genetic epidemiology level. Sci Rep 2019; 9:17583. [PMID: 31772290 PMCID: PMC6879509 DOI: 10.1038/s41598-019-53922-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 11/07/2019] [Indexed: 01/09/2023] Open
Abstract
We investigated the association between impulsivity related traits and BMI at the observational and genetic epidemiology level in a cross-sectional population of healthy young American-European adults. We studied 998 students and university staff of European ancestry recruited from Chicago (Illinois) and Athens (Georgia). We measured 14 impulsivity variables using three broad categories: impulsive choice, action and personality. Weight and height of participants were measured by research assistants. The single-nucleotide polymorphism (SNP) rs3751812 in the fat mass and obesity-associated (FTO) gene was genotyped using the Illumina PsychArray BeadChip platform. Within the three broad domains of impulsivity, 4 parameters (delay discounting of rewards area under the curve and average of k indexes, Conner's continuous performance test, and negative urgency) were associated with BMI. The FTO rs3751812 minor allele T was associated with higher BMI. Of the 14 impulsivity variables, rs3751812 T was associated with more premeditation and perseverance, before and after adjusting for BMI. The association between FTO rs3751812 and BMI adjusted for premeditation remained significant, but disappeared after adjusting for perseverance and for both perseverance and premeditation traits. Our observational and genetic data indicate a complex pattern of association between impulsive behaviors and BMI in healthy young American-European adults.
Collapse
Affiliation(s)
- David Meyre
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada.
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada.
| | - Sebat Mohamed
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Joshua C Gray
- Department of Medical and Clinical Psychology, Uniformed Services University, Bethesda, Maryland, United States of America
| | - Jessica Weafer
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, Illinois, United States of America
| | - James MacKillop
- Peter Boris Centre for Addictions Research, McMaster University/St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada.
| | - Harriet de Wit
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
28
|
Saber-Ayad M, Manzoor S, Radwan H, Hammoudeh S, Wardeh R, Ashraf A, Jabbar H, Hamoudi R. The FTO genetic variants are associated with dietary intake and body mass index amongst Emirati population. PLoS One 2019; 14:e0223808. [PMID: 31622411 PMCID: PMC6797190 DOI: 10.1371/journal.pone.0223808] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 09/27/2019] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND The risk of obesity is determined by complex interactions between genetic and environmental factors. Little research to date has investigated the interaction between gene and food intake. The aim of the current study is to explore the potential effect of fat mass and obesity-associated protein gene (FTO) rs9939609 and rs9930506 single nucleotide polymorphism (SNP) on the pattern of food intake in the Emirati population. METHODS Adult healthy Emirati subjects with Body mass index (BMI) of 16-40 kg/m2 were included in the study. Genotyping for FTO rs9939609(A>T) and rs9930506(A>G) was performed using DNA from saliva samples. Subjects were categorized according to the WHO classification by calculating the BMI to compare different classes. Dietary intake was assessed by a sixty-one-item FFQ that estimated food and beverage intakes over the past year. The daily energy, macronutrient, and micronutrient consumption were computed. RESULTS We included 169 subjects in the final analysis (mean age 30.49± 9.1years, 57.4% females). The mean BMI of the study population was 26.19 kg/m2. Both SNPs were in Hardy Weinberg Equilibrium. The rs9939609 AA genotype was significantly associated with higher BMI (p = 0.004); the effect was significant in females (p = 0.028), but not in males (p = 0.184). Carbohydrate intake was significantly higher in AA subjects with a trend of lower fat intake compared to other genotypes. The odds ratio for the AA was 3.78 in the fourth quartile and 2.67 for the A/T in the second quartile of total carbohydrate intake, considering the first quartile as a reference (95% CI = 1.017-14.1 and 1.03-6.88, respectively). Fat intake was significantly lower in the FTO rs9930506 GG subjects. The presence of FTO rs9930506 GG genotype decreased the fat intake in subjects with FTO rs9939609 AA (p = 0.037). CONCLUSIONS The results of this study highlight the interaction of the FTO risk alleles on the food intake in Emirati subjects. The FTO rs9939609 AA subjects had higher carbohydrate and lower fat intake. The latter was accentuated in presence of rs9930506 GG genotype.
Collapse
Affiliation(s)
- Maha Saber-Ayad
- College of Medicine, University of Sharjah, Sharjah, UAE
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, UAE
- College of Medicine, Cairo University, Cairo, Egypt
| | - Shaista Manzoor
- College of Medicine, University of Sharjah, Sharjah, UAE
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, UAE
| | - Hadia Radwan
- College of Medicine, University of Sharjah, Sharjah, UAE
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, UAE
- Clinical Nutrition and Dietetics Department, College of Health Sciences, University of Sharjah, Sharjah, UAE
| | - Sarah Hammoudeh
- College of Medicine, University of Sharjah, Sharjah, UAE
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, UAE
| | - Rahaf Wardeh
- College of Medicine, University of Sharjah, Sharjah, UAE
| | - Ahmed Ashraf
- College of Medicine, University of Sharjah, Sharjah, UAE
| | - Hussein Jabbar
- College of Medicine, University of Sharjah, Sharjah, UAE
| | - Rifat Hamoudi
- College of Medicine, University of Sharjah, Sharjah, UAE
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, UAE
| |
Collapse
|
29
|
Ashikawa Y, Shiromizu T, Miura K, Adachi Y, Matsui T, Bessho Y, Tanaka T, Nishimura Y. C3orf70 Is Involved in Neural and Neurobehavioral Development. Pharmaceuticals (Basel) 2019; 12:ph12040156. [PMID: 31623237 PMCID: PMC6958487 DOI: 10.3390/ph12040156] [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: 10/10/2019] [Accepted: 10/15/2019] [Indexed: 12/29/2022] Open
Abstract
Neurogenesis is the process by which undifferentiated progenitor cells develop into mature and functional neurons. Defects in neurogenesis are associated with neurodevelopmental and neuropsychiatric disorders; therefore, elucidating the molecular mechanisms underlying neurogenesis can advance our understanding of the pathophysiology of these disorders and facilitate the discovery of novel therapeutic targets. In this study, we performed a comparative transcriptomic analysis to identify common targets of the proneural transcription factors Neurog1/2 and Ascl1 during neurogenesis of human and mouse stem cells. We successfully identified C3orf70 as a novel common target gene of Neurog1/2 and Ascl1 during neurogenesis. Using in situ hybridization, we demonstrated that c3orf70a and c3orf70b, two orthologs of C3orf70, were expressed in the midbrain and hindbrain of zebrafish larvae. We generated c3orf70 knockout zebrafish using CRISPR/Cas9 technology and demonstrated that loss of c3orf70 resulted in significantly decreased expression of the mature neuron markers elavl3 and eno2. We also found that expression of irx3b, a zebrafish ortholog of IRX3 and a midbrain/hindbrain marker, was significantly reduced in c3orf70 knockout zebrafish. Finally, we demonstrated that neurobehaviors related to circadian rhythm and altered light–dark conditions were significantly impaired in c3orf70 knockout zebrafish. These results suggest that C3orf70 is involved in neural and neurobehavioral development and that defects in C3orf70 may be associated with midbrain/hindbrain-related neurodevelopmental and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Yoshifumi Ashikawa
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan; (Y.A.); (T.S.); (K.M.); (Y.A.)
| | - Takashi Shiromizu
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan; (Y.A.); (T.S.); (K.M.); (Y.A.)
| | - Koki Miura
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan; (Y.A.); (T.S.); (K.M.); (Y.A.)
| | - Yuka Adachi
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan; (Y.A.); (T.S.); (K.M.); (Y.A.)
| | - Takaaki Matsui
- Gene Regulation Research, Graduate School of Biological Sciences, Nara Institute of Science and Technology, Takayama, Nara 630-0192, Japan; (T.M.); (Y.B.)
| | - Yasumasa Bessho
- Gene Regulation Research, Graduate School of Biological Sciences, Nara Institute of Science and Technology, Takayama, Nara 630-0192, Japan; (T.M.); (Y.B.)
| | - Toshio Tanaka
- Department of Systems Pharmacology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan;
| | - Yuhei Nishimura
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan; (Y.A.); (T.S.); (K.M.); (Y.A.)
- Correspondence:
| |
Collapse
|
30
|
Doaei S, Kalantari N, Izadi P, Salonurmi T, Mosavi Jarrahi A, Rafieifar S, Azizi Tabesh G, Rahimzadeh G, Gholamalizadeh M, Goodarzi MO. Changes in FTO and IRX3 gene expression in obese and overweight male adolescents undergoing an intensive lifestyle intervention and the role of FTO genotype in this interaction. J Transl Med 2019; 17:176. [PMID: 31126299 PMCID: PMC6534854 DOI: 10.1186/s12967-019-1921-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 05/15/2019] [Indexed: 01/31/2023] Open
Abstract
Background Lifestyle intervention may have a critical effect on the association between genetics and obesity. This study aimed to investigate changes in FTO and IRX3 gene expression in obese and overweight male adolescents undergoing a lifestyle intervention and the role of FTO genotype in this interaction. Methods This study was a field trial of 62 adolescents from boys’ high schools in Tehran, Iran. Two schools were randomly allocated as the intervention (n = 30) and control (n = 32) schools. The rs9930506 SNP in FTO was genotyped at baseline and the level of FTO and IRX3 expression in peripheral blood mononuclear cells (PBMCs). Anthropometric measurements were assessed at baseline and after 18 weeks of intensive lifestyle intervention. Results Our results showed that IRX3 expression in the intervention group was significantly up-regulated compared to baseline (P = 0.007) and compared to the control group (P = 0.011).The intervention group had significantly up-regulated transcripts of IRX3 only in rs9930506 risk allele carriers of the intervention group compared to risk allele carriers of the control group (P = 0.017). Moreover, our data showed that the FTO expression was up-regulated in AA genotype carriers and down-regulated in AG/GG genotype carriers (P = 0.017). Conclusion Lifestyle modification may exert its effects on obesity through changes in the expression level of the FTO and IRX3 genes. However, FTO genotype plays a role in the extent of the effect of lifestyle changes on gene expression. Further studies are crucial to have a better understanding of the interaction between lifestyle, genetics and anthropometric measurements. Trial registration This paper reports a comprehensive intervention study (Interactions of Genetics, Lifestyle and Anthropometrics study or IGLA study), which is retrospectively registered in the Iranian Registry of Clinical Trials as IRCT2016020925699N2. Date registered: April 24, 2016. (https://www.irct.ir/searchresult.php?id=25699&number=2) Electronic supplementary material The online version of this article (10.1186/s12967-019-1921-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Saeid Doaei
- Research Center of Health and Environment, Guilan University of Medical Sciences, Rasht, Iran.,Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Naser Kalantari
- Department of Community Nutrition, School of Nutrition and Food Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Pantea Izadi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Tuire Salonurmi
- Department of Internal Medicine, Oulu University Hospital and University of Oulu, Oulu, Finland
| | | | - Shahram Rafieifar
- Health Promotion and Education Department, Ministry of Health, Tehran, Iran
| | - Ghasem Azizi Tabesh
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ghazaleh Rahimzadeh
- Institute for Intelligent Systems Research and Innovation (IISRI), Deakin University, Geelong Waurn Ponds, Australia
| | - Maryam Gholamalizadeh
- Student Research Committee, Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mark O Goodarzi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
31
|
Vasanth Rao VRB, Candasamy M, Bhattamisra SK. Obesity an overview: Genetic conditions and recent developments in therapeutic interventions. Diabetes Metab Syndr 2019; 13:2112-2120. [PMID: 31235145 DOI: 10.1016/j.dsx.2019.05.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 05/06/2019] [Indexed: 01/22/2023]
Abstract
Obesity is a complex disorder that is linked to many coexisting disorders. Recent epidemiological data have suggested that the prevalence of obesity is at an all-time high, growing to be one of the world's biggest problems. There are several mechanisms on how individuals develop obesity which includes genetic and environmental factors. Not only does obesity contribute to other health issues but it also greatly affects the quality of life, physical ability, mental strength and imposes a huge burden in terms of healthcare costs. Along with that, obesity is associated with the risk of mortality and has been shown to reduce the median survival rate. Obesity is basically when the body is not able to balance energy intake and output. When energy intake exceeds energy expenditure, excess calories will be stored as fat leading to weight gain and eventually obesity. The therapeutic market for treating obesity is composed of many different interventions from lifestyle intervention, surgical procedures to pharmacotherapeutic approaches. All of these interventions have their respective benefits and disadvantages and are specifically prescribed to a patient based on the severity of their obesity as well as the existence of other health conditions. This review discusses the genetic and environmental causes of obesity along with the recent developments in anti-obesity therapies.
Collapse
Affiliation(s)
- Vikram Rao B Vasanth Rao
- School of Postgraduate Studies, International Medical University, No 126, Jalan Jalil Perkasa 19, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia.
| | - Mayuren Candasamy
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Subrat Kumar Bhattamisra
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia.
| |
Collapse
|
32
|
Wang L, De Solis AJ, Goffer Y, Birkenbach KE, Engle SE, Tanis R, Levenson JM, Li X, Rausch R, Purohit M, Lee JY, Tan J, De Rosa MC, Doege CA, Aaron HL, Martins GJ, Brüning JC, Egli D, Costa R, Berbari N, Leibel RL, Stratigopoulos G. Ciliary gene RPGRIP1L is required for hypothalamic arcuate neuron development. JCI Insight 2019; 4:e123337. [PMID: 30728336 PMCID: PMC6413800 DOI: 10.1172/jci.insight.123337] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 01/03/2019] [Indexed: 12/16/2022] Open
Abstract
Intronic polymorphisms in the α-ketoglutarate-dependent dioxygenase gene (FTO) that are highly associated with increased body weight have been implicated in the transcriptional control of a nearby ciliary gene, retinitis pigmentosa GTPase regulator-interacting protein-1 like (RPGRIP1L). Previous studies have shown that congenital Rpgrip1l hypomorphism in murine proopiomelanocortin (Pomc) neurons causes obesity by increasing food intake. Here, we show by congenital and adult-onset Rpgrip1l deletion in Pomc-expressing neurons that the hyperphagia and obesity are likely due to neurodevelopmental effects that are characterized by a reduction in the Pomc/Neuropeptide Y (Npy) neuronal number ratio and marked increases in arcuate hypothalamic-paraventricular hypothalamic (ARH-PVH) axonal projections. Biallelic RPGRIP1L mutations result in fewer cilia-positive human induced pluripotent stem cell-derived (iPSC-derived) neurons and blunted responses to Sonic Hedgehog (SHH). Isogenic human ARH-like embryonic stem cell-derived (ESc-derived) neurons homozygous for the obesity-risk alleles at rs8050136 or rs1421085 have decreased RPGRIP1L expression and have lower numbers of POMC neurons. RPGRIP1L overexpression increases POMC cell number. These findings suggest that apparently functional intronic polymorphisms affect hypothalamic RPGRIP1L expression and impact development of POMC neurons and their derivatives, leading to hyperphagia and increased adiposity.
Collapse
Affiliation(s)
- Liheng Wang
- Naomi Berrie Diabetes Center and Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Alain J. De Solis
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Yossef Goffer
- Naomi Berrie Diabetes Center & Division of Molecular Genetics, Department of Pediatrics, College of Physicians and Surgeons of Columbia University, New York, New York, USA
| | - Kathryn E. Birkenbach
- Naomi Berrie Diabetes Center & Division of Molecular Genetics, Department of Pediatrics, College of Physicians and Surgeons of Columbia University, New York, New York, USA
| | - Staci E. Engle
- Department of Biology, Indiana University-Purdue University, Indianapolis, Indiana, USA
| | - Ross Tanis
- University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Jacob M. Levenson
- University of Central Florida College of Medicine, Orlando, Florida, USA
| | - Xueting Li
- Institute of Human Nutrition graduate program, Columbia University, New York, New York, USA
| | - Richard Rausch
- Naomi Berrie Diabetes Center & Division of Molecular Genetics, Department of Pediatrics, College of Physicians and Surgeons of Columbia University, New York, New York, USA
| | - Manika Purohit
- Zuckerman Institute, Columbia University, New York, New York, USA
| | - Jen-Yi Lee
- Cancer Research Laboratory Molecular Imaging Center, University of California, Berkeley, 94720, USA
| | - Jerica Tan
- Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Maria Caterina De Rosa
- Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Claudia A. Doege
- Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Holly L. Aaron
- Cancer Research Laboratory Molecular Imaging Center, University of California, Berkeley, 94720, USA
| | | | - Jens C. Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- National Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Dieter Egli
- Naomi Berrie Diabetes Center & Division of Molecular Genetics, Department of Pediatrics, College of Physicians and Surgeons of Columbia University, New York, New York, USA
| | - Rui Costa
- Zuckerman Institute, Columbia University, New York, New York, USA
| | - Nicolas Berbari
- Department of Biology, Indiana University-Purdue University, Indianapolis, Indiana, USA
| | - Rudolph L. Leibel
- Naomi Berrie Diabetes Center & Division of Molecular Genetics, Department of Pediatrics, College of Physicians and Surgeons of Columbia University, New York, New York, USA
| | - George Stratigopoulos
- Naomi Berrie Diabetes Center & Division of Molecular Genetics, Department of Pediatrics, College of Physicians and Surgeons of Columbia University, New York, New York, USA
| |
Collapse
|
33
|
Schneeberger M. Irx3, a new leader on obesity genetics. EBioMedicine 2018; 39:19-20. [PMID: 30541683 PMCID: PMC6354703 DOI: 10.1016/j.ebiom.2018.12.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 12/04/2018] [Indexed: 01/25/2023] Open
Affiliation(s)
- Marc Schneeberger
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA; KAVLI Neuroscience Institute Fellow, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|