1
|
Hjort L, Bredgaard SS, Manitta E, Marques I, Sørensen AE, Martino D, Grunnet LG, Kelstrup L, Houshmand-Oeregaard A, Clausen TD, Mathiesen ER, Olsen SF, Saffery R, Barrès R, Damm P, Vaag AA, Dalgaard LT. Epigenetics of the non-coding RNA nc886 across blood, adipose tissue and skeletal muscle in offspring exposed to diabetes in pregnancy. Clin Epigenetics 2024; 16:61. [PMID: 38715048 PMCID: PMC11077860 DOI: 10.1186/s13148-024-01673-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/20/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Diabetes in pregnancy is associated with increased risk of long-term metabolic disease in the offspring, potentially mediated by in utero epigenetic variation. Previously, we identified multiple differentially methylated single CpG sites in offspring of women with gestational diabetes mellitus (GDM), but whether stretches of differentially methylated regions (DMRs) can also be identified in adolescent GDM offspring is unknown. Here, we investigate which DNA regions in adolescent offspring are differentially methylated in blood by exposure to diabetes in pregnancy. The secondary aim was to characterize the RNA expression of the identified DMR, which contained the nc886 non-coding RNA. METHODS To identify DMRs, we employed the bump hunter method in samples from young (9-16 yr, n = 92) offspring of women with GDM (O-GDM) and control offspring (n = 94). Validation by pyrosequencing was performed in an adult offspring cohort (age 28-33 years) consisting of O-GDM (n = 82), offspring exposed to maternal type 1 diabetes (O-T1D, n = 67) and control offspring (O-BP, n = 57). RNA-expression was measured using RT-qPCR in subcutaneous adipose tissue and skeletal muscle. RESULTS One significant DMR represented by 10 CpGs with a bimodal methylation pattern was identified, located in the nc886/VTRNA2-1 non-coding RNA gene. Low methylation status across all CpGs of the nc886 in the young offspring was associated with maternal GDM. While low methylation degree in adult offspring in blood, adipose tissue, and skeletal muscle was not associated with maternal GDM, adipose tissue nc886 expression was increased in O-GDM compared to O-BP, but not in O-T1D. In addition, adipose tissue nc886 expression levels were positively associated with maternal pre-pregnancy BMI (p = 0.006), but not with the offspring's own adiposity. CONCLUSIONS Our results highlight that nc886 is a metastable epiallele, whose methylation in young offspring is negatively correlated with maternal obesity and GDM status. The physiological effect of nc886 may be more important in adipose tissue than in skeletal muscle. Further research should aim to investigate how nc886 regulation in adipose tissue by exposure to GDM may contribute to development of metabolic disease.
Collapse
Affiliation(s)
- Line Hjort
- Novo Nordisk Foundation Center for Basic Metabolic Research, Metabolic Epigenetics Group, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
- Center for Pregnant Women With Diabetes, Department of Obstetrics, Rigshospitalet, Copenhagen, Denmark.
| | | | - Eleonora Manitta
- Novo Nordisk Foundation Center for Basic Metabolic Research, Metabolic Epigenetics Group, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Irene Marques
- Center for Pregnant Women With Diabetes, Department of Obstetrics, Rigshospitalet, Copenhagen, Denmark
| | | | - David Martino
- Murdoch Children's Research Institute, Parkville, Melbourne, VIC, Australia
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Perth Children's Hospital, Nedlands, WA, Australia
| | - Louise Groth Grunnet
- Clinical Research, Steno Diabetes Center Copenhagen, Herlev Hospital, Herlev, Denmark
| | - Louise Kelstrup
- Center for Pregnant Women With Diabetes, Department of Obstetrics, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Gynecology and Obstetrics, Herlev Hospital, Herlev, Denmark
| | - Azadeh Houshmand-Oeregaard
- Center for Pregnant Women With Diabetes, Department of Obstetrics, Rigshospitalet, Copenhagen, Denmark
- Novo Nordisk A/S, Bagsværd, Denmark
| | - Tine Dalsgaard Clausen
- Center for Pregnant Women With Diabetes, Department of Obstetrics, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Elisabeth Reinhardt Mathiesen
- Center for Pregnant Women With Diabetes, Department of Obstetrics, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Endocrinology, Rigshospitalet, Copenhagen, Denmark
| | | | - Richard Saffery
- Murdoch Children's Research Institute, Parkville, Melbourne, VIC, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Romain Barrès
- Novo Nordisk Foundation Center for Basic Metabolic Research, Metabolic Epigenetics Group, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Damm
- Center for Pregnant Women With Diabetes, Department of Obstetrics, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Allan Arthur Vaag
- Clinical Research, Steno Diabetes Center Copenhagen, Herlev Hospital, Herlev, Denmark
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | | |
Collapse
|
2
|
Ling C, Bacos K, Rönn T. Epigenetics of type 2 diabetes mellitus and weight change - a tool for precision medicine? Nat Rev Endocrinol 2022; 18:433-448. [PMID: 35513492 DOI: 10.1038/s41574-022-00671-w] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/29/2022] [Indexed: 12/12/2022]
Abstract
Pioneering studies performed over the past few decades demonstrate links between epigenetics and type 2 diabetes mellitus (T2DM), the metabolic disorder with the most rapidly increasing prevalence in the world. Importantly, these studies identified epigenetic modifications, including altered DNA methylation, in pancreatic islets, adipose tissue, skeletal muscle and the liver from individuals with T2DM. As non-genetic factors that affect the risk of T2DM, such as obesity, unhealthy diet, physical inactivity, ageing and the intrauterine environment, have been associated with epigenetic modifications in healthy individuals, epigenetics probably also contributes to T2DM development. In addition, genetic factors associated with T2DM and obesity affect the epigenome in human tissues. Notably, causal mediation analyses found DNA methylation to be a potential mediator of genetic associations with metabolic traits and disease. In the past few years, translational studies have identified blood-based epigenetic markers that might be further developed and used for precision medicine to help patients with T2DM receive optimal therapy and to identify patients at risk of complications. This Review focuses on epigenetic mechanisms in the development of T2DM and the regulation of body weight in humans, with a special focus on precision medicine.
Collapse
Affiliation(s)
- Charlotte Ling
- Epigenetics and Diabetes Unit, Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, Scania University Hospital, Malmö, Sweden.
| | - Karl Bacos
- Epigenetics and Diabetes Unit, Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, Scania University Hospital, Malmö, Sweden
| | - Tina Rönn
- Epigenetics and Diabetes Unit, Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, Scania University Hospital, Malmö, Sweden
| |
Collapse
|
3
|
Xia K, Yu LY, Huang XQ, Zhao ZH, Liu J. Epigenetic regulation by long noncoding RNAs in osteo-/adipogenic differentiation of mesenchymal stromal cells and degenerative bone diseases. World J Stem Cells 2022; 14:92-103. [PMID: 35126830 PMCID: PMC8788182 DOI: 10.4252/wjsc.v14.i1.92] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 07/07/2021] [Accepted: 01/05/2022] [Indexed: 02/06/2023] Open
Abstract
Bone is a complex tissue that undergoes constant remodeling to maintain homeostasis, which requires coordinated multilineage differentiation and proper proliferation of mesenchymal stromal cells (MSCs). Mounting evidence indicates that a disturbance of bone homeostasis can trigger degenerative bone diseases, including osteoporosis and osteoarthritis. In addition to conventional genetic modifications, epigenetic modifications (i.e., DNA methylation, histone modifications, and the expression of noncoding RNAs) are considered to be contributing factors that affect bone homeostasis. Long noncoding RNAs (lncRNAs) were previously regarded as ‘transcriptional noise’ with no biological functions. However, substantial evidence suggests that lncRNAs have roles in the epigenetic regulation of biological processes in MSCs and related diseases. In this review, we summarized the interactions between lncRNAs and epigenetic modifiers associated with osteo-/adipogenic differentiation of MSCs and the pathogenesis of degenerative bone diseases and highlighted promising lncRNA-based diagnostic and therapeutic targets for bone diseases.
Collapse
Affiliation(s)
- Kai Xia
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Li-Yuan Yu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Xin-Qi Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Zhi-He Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Jun Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
4
|
Mirzaeicheshmeh E, Zerrweck C, Centeno-Cruz F, Baca-Peynado P, Martinez-Hernandez A, García-Ortiz H, Contreras-Cubas C, Salas-Martínez MG, Saldaña-Alvarez Y, Mendoza-Caamal EC, Barajas-Olmos F, Orozco L. Alterations of DNA methylation during adipogenesis differentiation of mesenchymal stem cells isolated from adipose tissue of patients with obesity is associated with type 2 diabetes. Adipocyte 2021; 10:493-504. [PMID: 34699309 PMCID: PMC8555535 DOI: 10.1080/21623945.2021.1978157] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 11/22/2022] Open
Abstract
Adipogenesis regulation is crucial for mature adipocyte function. In obesity, a major driver of type 2 diabetes (T2D), this process is disrupted and remains poorly characterized. Here we identified altered DNA methylation profiles in diabetic obese patients, during three adipocytes differentiation stages. We isolated mesenchymal cells from visceral adipose tissue of obese patients with and without T2D to analyse DNA methylation profiles at 0, 3, and 18 days of ex vivo differentiation and documented their impact on gene expression. Methylation and gene expression were analysed with EPIC and Clarion S arrays, respectively. Patients with T2D had epigenetic alterations in all the analysed stages, and these were mainly observed in genes important in adipogenesis, insulin resistance, cell death programming, and immune effector processes. Importantly, at 3 days, we found six-fold more methylated CpG alterations than in the other stages. This is the first study to document epigenetic markers that persist through all three adipogenesis stages and their impact on gene expression, which could be a cellular metabolic memory involved in T2D. Our data provided evidence that, throughout the adipogenesis process, alterations occur in methylation that might impact mature adipocyte function, cause tissue malfunction, and potentially, lead to the development of T2D.
Collapse
Affiliation(s)
- Elaheh Mirzaeicheshmeh
- Immunogenomics and Metabolic Disease Laboratory, Instituto Nacional De Medicina Genómica, Ss, Mexico City, Mexico
| | - Carlos Zerrweck
- Clínica de Obesidad del Hospital General Tláhuac, SSA, Mexico City, Mexico
- Facultad De Medicina, Alta Especialidad En Cirugía Bariatrica, Unam, Mexico City, Mexico
| | - Federico Centeno-Cruz
- Immunogenomics and Metabolic Disease Laboratory, Instituto Nacional De Medicina Genómica, Ss, Mexico City, Mexico
| | - Paulina Baca-Peynado
- Immunogenomics and Metabolic Disease Laboratory, Instituto Nacional De Medicina Genómica, Ss, Mexico City, Mexico
| | - Angélica Martinez-Hernandez
- Immunogenomics and Metabolic Disease Laboratory, Instituto Nacional De Medicina Genómica, Ss, Mexico City, Mexico
| | - Humberto García-Ortiz
- Immunogenomics and Metabolic Disease Laboratory, Instituto Nacional De Medicina Genómica, Ss, Mexico City, Mexico
| | - Cecilia Contreras-Cubas
- Immunogenomics and Metabolic Disease Laboratory, Instituto Nacional De Medicina Genómica, Ss, Mexico City, Mexico
| | | | - Yolanda Saldaña-Alvarez
- Immunogenomics and Metabolic Disease Laboratory, Instituto Nacional De Medicina Genómica, Ss, Mexico City, Mexico
| | | | - Francisco Barajas-Olmos
- Immunogenomics and Metabolic Disease Laboratory, Instituto Nacional De Medicina Genómica, Ss, Mexico City, Mexico
| | - Lorena Orozco
- Immunogenomics and Metabolic Disease Laboratory, Instituto Nacional De Medicina Genómica, Ss, Mexico City, Mexico
| |
Collapse
|
5
|
Shiu BH, Lu WY, Tantoh DM, Chou MC, Nfor ON, Huang CC, Liaw YP. Interactive association between dietary fat and sex on CDH13 cg02263260 methylation. BMC Med Genomics 2021; 14:13. [PMID: 33407434 PMCID: PMC7788866 DOI: 10.1186/s12920-020-00858-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 12/14/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND DNA methylation of Cadherin 13 (CDH13), a tumor suppressor gene is associated with gene repression and carcinogenesis. We determined the relation of dietary fat and sex with CDH13 cg02263260 methylation in Taiwanese adults. METHODS Data of 870 eligible participants (430 men and 440 women) between 30 and 70 years were obtained from the Taiwan Biobank (TWB) database. The association of dietary fat and sex with CDH13 cg02263260 methylation was determined using multiple linear regression. RESULTS The association between sex and cg02263260 methylation was significant: beta-coefficient (β) = 0.00532; 95% confidence interval (CI) = 0.00195-0.00868. Moreover, the interaction between sex and dietary fat on cg02263260 methylation was significant (P-value = 0.0145). After stratification by sex, the association of dietary fat with cg02263260 methylation was significant only in women. Specifically, high dietary fat was positively associated with cg02263260 methylation in women (β = 0.00597; 95% CI = 0.00061-0.01133) and the test for trend was significant (P-value = 0.0283). CONCLUSION High fat intake was significantly associated with higher cg02263260 methylation in women and the test for trend was significant. These findings suggest that the association of fat intake and CDH13 cg02263260 might vary by sex and CDH13 cg02263260 methylation levels in women might increase as fat intake increases.
Collapse
Affiliation(s)
- Bei-Hao Shiu
- Institute of Medicine, Chung Shan Medical University, Taichung City, 40201, Taiwan
- Division of Colon-Rectal Surgery, Department of Surgery, Chung Shan Medical University Hospital, Taichung City, 40201, Taiwan
| | - Wen-Yu Lu
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, No. 110 Sec. 1 Jianguo N. Road, Taichung City, 40201, Taiwan
| | - Disline Manli Tantoh
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, No. 110 Sec. 1 Jianguo N. Road, Taichung City, 40201, Taiwan
- Department of Medical Imaging, Chung Shan Medical University Hospital, Taichung City, 40201, Taiwan
| | - Ming-Chih Chou
- Institute of Medicine, Chung Shan Medical University, Taichung City, 40201, Taiwan
| | - Oswald Ndi Nfor
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, No. 110 Sec. 1 Jianguo N. Road, Taichung City, 40201, Taiwan
| | - Chi-Chou Huang
- Division of Colon-Rectal Surgery, Department of Surgery, Chung Shan Medical University Hospital, Taichung City, 40201, Taiwan.
- School of Medicine, Chung Shan Medical University, No. 110 Sec. 1 Jianguo N. Road, Taichung City, 40201, Taiwan.
| | - Yung-Po Liaw
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, No. 110 Sec. 1 Jianguo N. Road, Taichung City, 40201, Taiwan.
- Department of Medical Imaging, Chung Shan Medical University Hospital, Taichung City, 40201, Taiwan.
| |
Collapse
|
6
|
Parrillo L, Spinelli R, Longo M, Desiderio A, Mirra P, Nigro C, Fiory F, Hedjazifar S, Mutarelli M, Carissimo A, Formisano P, Miele C, Smith U, Raciti GA, Beguinot F. Altered PTPRD DNA methylation associates with restricted adipogenesis in healthy first-degree relatives of Type 2 diabetes subjects. Epigenomics 2020; 12:873-888. [PMID: 32483983 DOI: 10.2217/epi-2019-0267] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aim: First-degree relatives (FDR) of individuals with Type 2 diabetes (T2D) feature restricted adipogenesis, which render them more vulnerable to T2D. Epigenetics may contribute to these abnormalities. Methods: FDR pre-adipocyte Methylome and Transcriptome were investigated by MeDIP- and RNA-Seq, respectively. Results: Methylome analysis revealed 2841 differentially methylated regions (DMR) in FDR. Most DMR localized into gene-body and were hypomethylated. The strongest hypomethylation signal was identified in an intronic-DMR at the PTPRD gene. PTPRD hypomethylation in FDR was confirmed by bisulphite sequencing and was responsible for its upregulation. Interestingly, Ptprd-overexpression in 3T3-L1 pre-adipocytes inhibited adipogenesis. Notably, the validated PTPRD-associated DMR was significantly hypomethylated in peripheral blood leukocytes from the same FDR individuals. Finally, PTPRD methylation pattern was also replicated in obese individuals. Conclusion: Our findings indicated a previously unrecognized role of PTPRD in restraining adipogenesis. This abnormality may contribute to increase FDR proclivity toward T2D.
Collapse
Affiliation(s)
- Luca Parrillo
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine - Federico II University of Naples, 80131, Italy
| | - Rosa Spinelli
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine - Federico II University of Naples, 80131, Italy
| | - Michele Longo
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine - Federico II University of Naples, 80131, Italy
| | - Antonella Desiderio
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine - Federico II University of Naples, 80131, Italy
| | - Paola Mirra
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine - Federico II University of Naples, 80131, Italy
| | - Cecilia Nigro
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine - Federico II University of Naples, 80131, Italy
| | - Francesca Fiory
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine - Federico II University of Naples, 80131, Italy
| | - Shahram Hedjazifar
- Lundberg Laboratory for Diabetes Research, Department of Molecular & Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 41345, Sweden
| | | | | | - Pietro Formisano
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine - Federico II University of Naples, 80131, Italy
| | - Claudia Miele
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine - Federico II University of Naples, 80131, Italy
| | - Ulf Smith
- Lundberg Laboratory for Diabetes Research, Department of Molecular & Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 41345, Sweden
| | - Gregory Alexander Raciti
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine - Federico II University of Naples, 80131, Italy
| | - Francesco Beguinot
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine - Federico II University of Naples, 80131, Italy
| |
Collapse
|
7
|
Zhang M, Li D, Zhai Y, Wang Z, Ma X, Zhang D, Li G, Han R, Jiang R, Li Z, Kang X, Sun G. The Landscape of DNA Methylation Associated With the Transcriptomic Network of Intramuscular Adipocytes Generates Insight Into Intramuscular Fat Deposition in Chicken. Front Cell Dev Biol 2020; 8:206. [PMID: 32300590 PMCID: PMC7142253 DOI: 10.3389/fcell.2020.00206] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 03/10/2020] [Indexed: 12/13/2022] Open
Abstract
Intramuscular fat (IMF), which regulated by genetics, nutrition and environment is an important factor that influencing meat quality. Up to now, the epigenetic regulation mechanism underlying poultry IMF deposition remains poorly understood. Here, we focused on the DNA methylation, which usually regulate genes in transcription level. To look into the essential role of DNA methylation on the IMF deposition, chicken intramuscular preadipocytes were isolated and cultured in vitro, and a model of intramuscular adipocyte differentiation was constructed. Combined the whole genome bisulfite sequencing (WGBS) and RNA-Seq technologies, we identified several methylated genes, which mainly affecting fatty acid metabolism and muscle development. Furthermore, we reported that DNA methylation regulate intramuscular adipogenesis by regulating the genes, such as collagen, type VI, alpha 1 (COL6A1) thus affecting IMF deposition. Overexpression of COL6A1 increases the lipid droplet and inhibits cell proliferation by regulating CHAD and CAMK2 in intramuscular adipocytes, while knockdown of COL6A1 shows the opposite effect. Taken together, our results reveal that DNA methylation plays an important role in poultry IMF deposition.
Collapse
Affiliation(s)
- Meng Zhang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Donghua Li
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Yanhui Zhai
- The First Clinical Hospital, Jilin University, Changchun, China
| | - Zhengzhu Wang
- The First Clinical Hospital, Jilin University, Changchun, China
| | - Xiangfei Ma
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Daoyu Zhang
- The First Clinical Hospital, Jilin University, Changchun, China
| | - Guoxi Li
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Ruili Han
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Ruirui Jiang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Zhuanjian Li
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Xiangtao Kang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Guirong Sun
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| |
Collapse
|
8
|
Stachecka J, Lemanska W, Noak M, Szczerbal I. Expression of key genes involved in DNA methylation during in vitro differentiation of porcine mesenchymal stem cells (MSCs) into adipocytes. Biochem Biophys Res Commun 2020; 522:811-818. [DOI: 10.1016/j.bbrc.2019.11.175] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 11/26/2019] [Indexed: 12/12/2022]
|
9
|
Zhang Q, Nettleship I, Schmelzer E, Gerlach J, Zhang X, Wang J, Liu C. Tissue Engineering and Regenerative Medicine Therapies for Cell Senescence in Bone and Cartilage. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:64-78. [DOI: 10.1089/ten.teb.2019.0215] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Qinghao Zhang
- Department of Materials Science and Engineering, East China University of Science and Technology, Shanghai, P.R. China
- Department of Mechanical Engineering and Materials Science, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ian Nettleship
- Department of Mechanical Engineering and Materials Science, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Eva Schmelzer
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jorg Gerlach
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Xuewei Zhang
- Department of Materials Science and Engineering, East China University of Science and Technology, Shanghai, P.R. China
| | - Jing Wang
- Department of Materials Science and Engineering, East China University of Science and Technology, Shanghai, P.R. China
| | - Changsheng Liu
- Department of Materials Science and Engineering, East China University of Science and Technology, Shanghai, P.R. China
| |
Collapse
|
10
|
Transgenerational Self-Reconstruction of Disrupted Chromatin Organization After Exposure To An Environmental Stressor in Mice. Sci Rep 2019; 9:13057. [PMID: 31506492 PMCID: PMC6736928 DOI: 10.1038/s41598-019-49440-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 08/21/2019] [Indexed: 02/07/2023] Open
Abstract
Exposure to environmental stressors is known to increase disease susceptibility in unexposed descendants in the absence of detectable genetic mutations. The mechanisms mediating environmentally-induced transgenerational disease susceptibility are poorly understood. We showed that great-great-grandsons of female mice exposed to tributyltin (TBT) throughout pregnancy and lactation were predisposed to obesity due to altered chromatin organization that subsequently biased DNA methylation and gene expression. Here we analyzed DNA methylomes and transcriptomes from tissues of animals ancestrally exposed to TBT spanning generations, sexes, ontogeny, and cell differentiation state. We found that TBT elicited concerted alterations in the expression of “chromatin organization” genes and inferred that TBT-disrupted chromatin organization might be able to self-reconstruct transgenerationally. We also found that the location of “chromatin organization” and “metabolic” genes is biased similarly in mouse and human genomes, suggesting that exposure to environmental stressors in different species could elicit similar phenotypic effects via self-reconstruction of disrupted chromatin organization.
Collapse
|
11
|
Samblas M, Milagro FI, Martínez A. DNA methylation markers in obesity, metabolic syndrome, and weight loss. Epigenetics 2019; 14:421-444. [PMID: 30915894 DOI: 10.1080/15592294.2019.1595297] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The fact that not all individuals exposed to the same environmental risk factors develop obesity supports the hypothesis of the existence of underlying genetic and epigenetic elements. There is suggestive evidence that environmental stimuli, such as dietary pattern, particularly during pregnancy and early life, but also in adult life, can induce changes in DNA methylation predisposing to obesity and related comorbidities. In this context, the DNA methylation marks of each individual have emerged not only as a promising tool for the prediction, screening, diagnosis, and prognosis of obesity and metabolic syndrome features, but also for the improvement of weight loss therapies in the context of precision nutrition. The main objectives in this field are to understand the mechanisms involved in transgenerational epigenetic inheritance, and featuring the nutritional and lifestyle factors implicated in the epigenetic modifications. Likewise, DNA methylation modulation caused by diet and environment may be a target for newer therapeutic strategies concerning the prevention and treatment of metabolic diseases.
Collapse
Affiliation(s)
- Mirian Samblas
- a Department of Nutrition, Food Science and Physiology; Centre for Nutrition Research , University of Navarra , Pamplona , Spain
| | - Fermín I Milagro
- a Department of Nutrition, Food Science and Physiology; Centre for Nutrition Research , University of Navarra , Pamplona , Spain.,b CIBERobn, CIBER Fisiopatología de la Obesidad y Nutrición , Instituto de Salud Carlos III. Madrid , Spain.,c IdiSNA, Instituto de Investigación Sanitaria de Navarra (IdiSNA) , Pamplona , Spain
| | - Alfredo Martínez
- a Department of Nutrition, Food Science and Physiology; Centre for Nutrition Research , University of Navarra , Pamplona , Spain.,b CIBERobn, CIBER Fisiopatología de la Obesidad y Nutrición , Instituto de Salud Carlos III. Madrid , Spain.,c IdiSNA, Instituto de Investigación Sanitaria de Navarra (IdiSNA) , Pamplona , Spain.,d IMDEA, Research Institute on Food & Health Sciences , Madrid , Spain
| |
Collapse
|
12
|
Flores EM, Woeller CF, Falsetta ML, Susiarjo M, Phipps RP. Thy1 (CD90) expression is regulated by DNA methylation during adipogenesis. FASEB J 2019; 33:3353-3363. [PMID: 30376360 PMCID: PMC6404567 DOI: 10.1096/fj.201801481r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 10/09/2018] [Indexed: 12/23/2022]
Abstract
The obesity epidemic is developing into the most costly health problem facing the world. Obesity, characterized by excessive adipogenesis and enlarged adipocytes, promotes morbidities, such as diabetes, cardiovascular disease, and cancer. Regulation of adipogenesis is critical to our understanding of how fat cell formation causes obesity and associated health problems. Thy1 (also called CD90), a widely used stem cell marker, blocks adipogenesis and reduces lipid accumulation. Thy1-knockout mice are prone to diet-induced obesity. Although the importance of Thy1 in adipogenesis and obesity is now evident, how its expression is regulated is not. We hypothesized that DNA methylation has a role in promoting adipogenesis and affects Thy1 expression. Using the methylation inhibitor 5-aza-2'-deoxycytidine (5-aza-dC), we investigated whether DNA methylation alters Thy1 expression during adipogenesis in both mouse 3T3-L1 preadipocytes and mouse mesenchymal stem cells. Thy1 protein and mRNA levels were decreased dramatically during adipogenesis. However, 5-aza-dC treatment prevented that phenomenon. Methylation-sensitive pyrosequencing analysis showed that CpG sites at the Thy1 locus have increased methylation during adipogenesis, as well as increased methylation in adipose tissue from diet-induced obese mice. These new findings highlight the potential role of Thy1 and DNA methylation in adipogenesis and obesity.-Flores, E. M., Woeller, C. F., Falsetta, M. L., Susiarjo, M., Phipps, R. P. Thy1 (CD90) expression is regulated by DNA methylation during adipogenesis.
Collapse
Affiliation(s)
- E’Lissa M. Flores
- Clinical and Translational Science Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Collynn F. Woeller
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA; and
| | - Megan L. Falsetta
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA; and
| | - Martha Susiarjo
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA; and
| | - Richard P. Phipps
- Clinical and Translational Science Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA; and
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
13
|
Paradoxical effects of the epigenetic modifiers 5-aza-deoxycytidine and suberoylanilide hydroxamic acid on adipogenesis. Differentiation 2019; 106:1-8. [PMID: 30818187 DOI: 10.1016/j.diff.2019.02.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 01/12/2019] [Accepted: 02/13/2019] [Indexed: 01/17/2023]
Abstract
Adipogenesis is an important biological process that is linked to obesity and metabolic disorders. On the other hand, fat regeneration is crucial as a restorative approach following mastectomy or severe burn injury. Furthermore, optimizing an in-vitro model of adipogenesis, which would help in understanding the possible effects and/or side effects of fat-soluble drugs and anti-obesity remedies, in addition to the developmental studies. Epigenetic is an important factor that is involved in cellular differentiation and commitment. This study aimed at investigating the effect of DNA methylation and histone deactylases inhibitors, 5-Aza-deoxycytidine (5-Aza-dC) and Suberoylanilide hydroxamic acid (SAHA), on the adipogenic differentiation process. The two modifiers were applied according to our previously published protocol, followed by three cycles of a classical, two-step adipogenesis protocol. The cells pretreated with SAHA showed enhanced expression of the many adipogenic genes, including peroxisome proliferator-activated receptor-γ as well as the accumulation of intracytoplasmic fat as shown by oil red and Nile red staining and the secretion of adipokines, such as MCP-1 and IP-10. On contrary, 5-Aza-dC inhibited all these markers. In conclusion, adding the reported step with SAHA to the differentiation protocols could have an impact on the progress of the in-vitro fat regenerative approach. The possible role of 5-Aza-dC in the inhibition of adipogenesis can be of clinical interest and will need further characterization in the future.
Collapse
|
14
|
Ford SM, Simon Peter L, Berner P, Cook G, Vande Stouwe C, Dufour J, Bagby G, Nelson S, Molina PE. Differential contribution of chronic binge alcohol and antiretroviral therapy to metabolic dysregulation in SIV-infected male macaques. Am J Physiol Endocrinol Metab 2018; 315:E892-E903. [PMID: 30040479 PMCID: PMC6293168 DOI: 10.1152/ajpendo.00175.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/20/2018] [Accepted: 07/23/2018] [Indexed: 02/08/2023]
Abstract
The incidence of alcohol use disorder (AUD) is higher among people living with HIV (PLWH). The advent and continued development of antiretroviral therapy (ART) has significantly reduced mortality, shifting the course of HIV infection to a chronic illness. However, this is associated with an increased incidence of comorbid conditions, including type 2 diabetes mellitus, insulin resistance, and cardiovascular complications. Using a nonhuman primate model of simian immunodeficiency virus (SIV) infection, previous studies have demonstrated that chronic binge alcohol (CBA) administration decreases whole body insulin responsiveness, irrespective of ART administration. The objective of the current study was to determine the effects of CBA and ART on insulin-sensitive peripheral tissues before the development of overt clinical symptoms of SIV disease. Our results show that CBA reduced omental adipocyte cell size, increased collagen expression, and decreased the in vitro differentiation potential of adipose-derived stem cells. In contrast, it did not alter skeletal muscle or omental or hepatic expression of insulin signaling proteins. However, ART significantly decreased skeletal muscle expression of phosphatase and tensin homolog, total mechanistic target of rapamycin, and ribosomal protein S6. In addition, ART increased hepatic phosphorylation of AMP-activated protein kinase α and increased gene expression of key enzymes required for gluconeogenesis and fatty acid synthesis. These findings suggest that CBA and ART differentially promote adverse metabolic effects in an organ-specific manner that may underlie insulin resistance associated with alcohol, SIV, and ART. Whether this is translated in PLWH with AUD remains to be determined.
Collapse
Affiliation(s)
- Stephen M Ford
- Department of Physiology, Comprehensive Alcohol Research Center, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| | - Liz Simon Peter
- Department of Physiology, Comprehensive Alcohol Research Center, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| | - Paul Berner
- Department of Physiology, Comprehensive Alcohol Research Center, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| | - Garth Cook
- Department of Physiology, Comprehensive Alcohol Research Center, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| | - Curtis Vande Stouwe
- Department of Physiology, Comprehensive Alcohol Research Center, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| | - Jason Dufour
- Divison of Veterinary Medicine, Tulane National Primate Research Center , Covington, Louisiana
| | - Gregory Bagby
- Department of Physiology, Comprehensive Alcohol Research Center, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| | - Steve Nelson
- School of Medicine, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| | - Patricia E Molina
- Department of Physiology, Comprehensive Alcohol Research Center, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| |
Collapse
|
15
|
Tang J, Pulliam N, Özeş A, Buechlein A, Ding N, Keer H, Rusch D, O'Hagan H, Stack MS, Nephew KP. Epigenetic Targeting of Adipocytes Inhibits High-Grade Serous Ovarian Cancer Cell Migration and Invasion. Mol Cancer Res 2018; 16:1226-1240. [PMID: 29759990 PMCID: PMC6072573 DOI: 10.1158/1541-7786.mcr-17-0406] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 11/16/2017] [Accepted: 04/27/2018] [Indexed: 01/17/2023]
Abstract
Ovarian cancer (OC) cells frequently metastasize to the omentum, and adipocytes play a significant role in ovarian tumor progression. Therapeutic interventions targeting aberrant DNA methylation in ovarian tumors have shown promise in the clinic, but the effects of epigenetic therapy on the tumor microenvironment are understudied. Here, we examined the effect of adipocytes on OC cell behavior in culture and impact of targeting DNA methylation in adipocytes on OC metastasis. The presence of adipocytes increased OC cell migration and invasion, and proximal and direct coculture of adipocytes increased OC proliferation alone or after treatment with carboplatin. Treatment of adipocytes with hypomethylating agent guadecitabine decreased migration and invasion of OC cells toward adipocytes. Subcellular protein fractionation of adipocytes treated with guadecitabine revealed decreased DNA methyltransferase 1 (DNMT1) levels even in the presence of DNA synthesis inhibitor, aphidicolin. Methyl-Capture- and RNA-sequencing analysis of guadecitabine-treated adipocytes revealed derepression of tumor-suppressor genes and epithelial-mesenchymal transition inhibitors. SUSD2, a secreted tumor suppressor downregulated by promoter CpG island methylation in adipocytes, was upregulated after guadecitabine treatment, and recombinant SUSD2 decreased OC cell migration and invasion. Integrated analysis of the methylomic and transcriptomic data identified pathways associated with inhibition of matrix metalloproteases and fatty acid α-oxidation, suggesting a possible mechanism of how epigenetic therapy of adipocytes decreases metastasis. In conclusion, the effect of DNMT inhibitor on fully differentiated adipocytes suggests that hypomethylating agents may affect the tumor microenvironment to decrease cancer cell metastasis.Implications: Epigenetic targeting of tumor microenvironment can affect metastatic behavior of ovarian cancer cells. Mol Cancer Res; 16(8); 1226-40. ©2018 AACR.
Collapse
Affiliation(s)
- Jessica Tang
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana
| | - Nicholas Pulliam
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana
- Molecular and Cellular Biochemistry Department, Indiana University, Bloomington, Indiana
| | - Ali Özeş
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana
- Molecular and Cellular Biochemistry Department, Indiana University, Bloomington, Indiana
| | - Aaron Buechlein
- Center of Genomics and Bioinformatics, Indiana University, Bloomington, Indiana
| | - Ning Ding
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana
| | - Harold Keer
- Astex Pharmaceuticals Inc., Pleasanton, California
| | - Doug Rusch
- Center of Genomics and Bioinformatics, Indiana University, Bloomington, Indiana
| | - Heather O'Hagan
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
- Indiana University Simon Cancer Center, Indianapolis, Indiana
| | - M Sharon Stack
- Department of Chemistry and Biochemistry, Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana
| | - Kenneth P Nephew
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana.
- Molecular and Cellular Biochemistry Department, Indiana University, Bloomington, Indiana
- Indiana University Simon Cancer Center, Indianapolis, Indiana
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
16
|
Maurizi G, Babini L, Della Guardia L. Potential role of microRNAs in the regulation of adipocytes liposecretion and adipose tissue physiology. J Cell Physiol 2018; 233:9077-9086. [PMID: 29932216 DOI: 10.1002/jcp.26523] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 01/31/2018] [Indexed: 12/19/2022]
Abstract
Adipose tissue is a dynamic endocrine organ playing a pivotal role in metabolism modulation. Adipocytes differentiation requires a highly orchestrated series of changes of gene expression in precursor cells. At the same time, white mature adipocytes are plastic cells able to reversibly transdifferentiate toward fibroblast-like cells via the liposecretion process, returning back to a non-committed status of the cells. In particular, adipose tissue microenvironment along with external signaling molecules such as adipokines, cytokines and growth factors can regulate adipocytes physiology through complex molecular networks. MicroRNAs (miRNAs), a type of non-coding RNA, acting as fine regulators of biological processes and their expression is sensible to the environment and cellular status changes. MiRNAs are thought to play a pivotal role in regulating the physiology of adipose tissue as well as in the development of obesity and associated metabolic disturbances, although the underlying mechanisms have not been identified so far. Elucidating the molecular mechanisms orchestrating adipose tissue biology is required to better characterize obesity and its associated diseases. In this respect, the review aims to analyze the microRNAs potentially involved in adipogenesis highlighting their role in the process of liposecretion, adipocyte proliferation, and adipokines secretion. The role of microRNAs in the development of obesity and obesity-associated disorders is also discussed.
Collapse
Affiliation(s)
| | - Lucia Babini
- Università Politecnica delle Marche, Ancona, Italy
| | - Lucio Della Guardia
- Dipartimento di Sanità Pubblica, Medicina Sperimentale e Forense, Unità di Scienza dell'Alimentazione, Università degli studi di Pavia, Pavia, Italy
| |
Collapse
|
17
|
|
18
|
Maurizi G, Della Guardia L, Maurizi A, Poloni A. Adipocytes properties and crosstalk with immune system in obesity-related inflammation. J Cell Physiol 2017; 233:88-97. [PMID: 28181253 DOI: 10.1002/jcp.25855] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 02/07/2017] [Indexed: 12/11/2022]
Abstract
Obesity is a condition likely associated with several dysmetabolic conditions or worsening of cardiovascular and other chronic disturbances. A key role in this mechanism seem to be played by the onset of low-grade systemic inflammation, highlighting the importance of the interplay between adipocytes and immune system cells. Adipocytes express a complex and highly adaptive biological profile being capable to selectively activate different metabolic pathways in order to respond to environmental stimuli. It has been demonstrated how adipocytes, under appropriate stimulation, can easily differentiate and de-differentiate thereby converting themselves into different phenotypes according to metabolic necessities. Although underlying mechanisms are not fully understood, growing in adipocyte size and the inability of storing triglycerides under overfeeding conditions seem to be crucial for the switching to a dysfunctional metabolic profile, which is characterized by inflammatory and apoptotic pathways activation, and by the shifting to pro-inflammatory adipokines secretion. In obesity, changes in adipokines secretion along with adipocyte deregulation and fatty acids release into circulation contribute to maintain immune cells activation as well as their infiltration into regulatory organs. Over the well-established role of macrophages, recent findings suggest the involvement of new classes of immune cells such as T regulatory lymphocytes and neutrophils in the development inflammation and multi systemic worsening. Deeply understanding the pathways of adipocyte regulation and the de-differentiation process could be extremely useful for developing novel strategies aimed at curbing obesity-related inflammation and related metabolic disorders.
Collapse
Affiliation(s)
- Giulia Maurizi
- Clinica di Ematologia, Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, Italy
| | - Lucio Della Guardia
- Dipartimento di Sanità Pubblica, Medicina Sperimentale e Forense, Unità di Scienza dell'Alimentazione, Università degli studi di Pavia, Pavia, Italy
| | - Angela Maurizi
- Chirurgia d'Urgenza e del Trauma, Azienda Ospedaliera Universitaria-Ospedali Riuniti di Ancona, Ancona, Italy
| | - Antonella Poloni
- Clinica di Ematologia, Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|