1
|
Alsaedi S, Ogasawara M, Alarawi M, Gao X, Gojobori T. AI-powered precision medicine: utilizing genetic risk factor optimization to revolutionize healthcare. NAR Genom Bioinform 2025; 7:lqaf038. [PMID: 40330081 PMCID: PMC12051108 DOI: 10.1093/nargab/lqaf038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 02/11/2025] [Accepted: 04/17/2025] [Indexed: 05/08/2025] Open
Abstract
The convergence of artificial intelligence (AI) and biomedical data is transforming precision medicine by enabling the use of genetic risk factors (GRFs) for customized healthcare services based on individual needs. Although GRFs play an essential role in disease susceptibility, progression, and therapeutic outcomes, a gap exists in exploring their contribution to AI-powered precision medicine. This paper addresses this need by investigating the significance and potential of utilizing GRFs with AI in the medical field. We examine their applications, particularly emphasizing their impact on disease prediction, treatment personalization, and overall healthcare improvement. This review explores the application of AI algorithms to optimize the use of GRFs, aiming to advance precision medicine in disease screening, patient stratification, drug discovery, and understanding disease mechanisms. Through a variety of case studies and examples, we demonstrate the potential of incorporating GRFs facilitated by AI into medical practice, resulting in more precise diagnoses, targeted therapies, and improved patient outcomes. This review underscores the potential of GRFs, empowered by AI, to enhance precision medicine by improving diagnostic accuracy, treatment precision, and individualized healthcare solutions.
Collapse
Affiliation(s)
- Sakhaa Alsaedi
- Computer Science, Division of Computer, Electrical and Mathematical Sciences and Engineering (CEMSE), King Abdullah University of Science and Technology (KAUST), 23955-6900 Thuwal, Kingdom of Saudi Arabia
- Center of Excellence on Smart Health, King Abdullah University of Science and Technology (KAUST), 23955-6900 Thuwal, Kingdom of Saudi Arabia
- Center of Excellence for Generative AI, King Abdullah University of Science and Technology (KAUST), 23955-6900 Thuwal, Kingdom of Saudi Arabia
- College of Computer Science and Engineering (CCSE), Taibah University, 42353 Madinah, Kingdom of Saudi Arabia
| | - Michihiro Ogasawara
- Department of Internal Medicine and Rheumatology, Juntendo University, 113-8431 Tokyo, Japan
| | - Mohammed Alarawi
- Center of Excellence on Smart Health, King Abdullah University of Science and Technology (KAUST), 23955-6900 Thuwal, Kingdom of Saudi Arabia
- Center of Excellence for Generative AI, King Abdullah University of Science and Technology (KAUST), 23955-6900 Thuwal, Kingdom of Saudi Arabia
- Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology (KAUST), 23955-6900 Thuwal, Kingdom of Saudi Arabia
| | - Xin Gao
- Computer Science, Division of Computer, Electrical and Mathematical Sciences and Engineering (CEMSE), King Abdullah University of Science and Technology (KAUST), 23955-6900 Thuwal, Kingdom of Saudi Arabia
- Center of Excellence on Smart Health, King Abdullah University of Science and Technology (KAUST), 23955-6900 Thuwal, Kingdom of Saudi Arabia
- Center of Excellence for Generative AI, King Abdullah University of Science and Technology (KAUST), 23955-6900 Thuwal, Kingdom of Saudi Arabia
| | - Takashi Gojobori
- Center of Excellence on Smart Health, King Abdullah University of Science and Technology (KAUST), 23955-6900 Thuwal, Kingdom of Saudi Arabia
- Center of Excellence for Generative AI, King Abdullah University of Science and Technology (KAUST), 23955-6900 Thuwal, Kingdom of Saudi Arabia
- Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology (KAUST), 23955-6900 Thuwal, Kingdom of Saudi Arabia
- Marine Open Innovation Institute (MaOI), 113-8431 Shizuoka, Japan
| |
Collapse
|
2
|
Yashaswini C, Kiran NS, Chatterjee A. Zebrafish navigating the metabolic maze: insights into human disease - assets, challenges and future implications. J Diabetes Metab Disord 2025; 24:3. [PMID: 39697864 PMCID: PMC11649609 DOI: 10.1007/s40200-024-01539-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/26/2024] [Indexed: 12/20/2024]
Abstract
Zebrafish (Danio rerio) have become indispensable models for advancing our understanding of multiple metabolic disorders such as obesity, diabetes mellitus, dyslipidemia, and metabolic syndrome. This review provides a comprehensive analysis of zebrafish as a powerful tool for dissecting the genetic and molecular mechanisms of these diseases, focusing on key genes, like pparγ, lepr, ins, and srebp. Zebrafish offer distinct advantages, including genetic tractability, optical transparency in early development, and the conservation of key metabolic pathways with humans. Studies have successfully used zebrafish to uncover conserved metabolic mechanisms, identify novel disease pathways, and facilitate high-throughput screening of potential therapeutic compounds. The review also highlights the novelty of using zebrafish to model multifactorial metabolic disorders, addressing challenges such as interspecies differences in metabolism and the complexity of human metabolic disease etiology. Moving forward, future research will benefit from integrating advanced omics technologies to map disease-specific molecular signatures, applying personalized medicine approaches to optimize treatments, and utilizing computational models to predict therapeutic outcomes. By embracing these innovative strategies, zebrafish research has the potential to revolutionize the diagnosis, treatment, and prevention of metabolic disorders, offering new avenues for translational applications. Continued interdisciplinary collaboration and investment in zebrafish-based studies will be crucial to fully harnessing their potential for advancing therapeutic development.
Collapse
Affiliation(s)
- Chandrashekar Yashaswini
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru, Karnataka 560064 India
| | | | - Ankita Chatterjee
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru, Karnataka 560064 India
| |
Collapse
|
3
|
Huang C, Luo Y, Liu Y, Liu J, Chen Y, Zeng B, Liao X, Liu Y, Wang X. DNA hypermethylation-induced suppression of ALKBH5 is required for folic acid to alleviate hepatic lipid deposition by enhancing autophagy in an ATG12-dependent manner. J Nutr Biochem 2025; 140:109870. [PMID: 39993647 DOI: 10.1016/j.jnutbio.2025.109870] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 12/08/2024] [Accepted: 02/13/2025] [Indexed: 02/26/2025]
Abstract
Nonalcoholic fatty liver disease (NAFLD) occurs when too much fat builds up in the liver. As a growing worldwide epidemic, NAFLD is strongly linked with multiple metabolic diseases including obesity, insulin resistance, and dyslipidemia. However, very few effective treatments are currently available. Folate, an essential B-group vitamin with important biological functions including DNA and RNA methylation regulation, has been shown to have a protective effect against NAFLD with its underlying mechanism remains largely unclear. Here, we show that administration of folic acid significantly improves glucose tolerance, insulin sensitivity, and dyslipidemia in high-fat diet (HFD) fed mice. Moreover, folic acid treatment significantly inhibits lipid deposition in hepatocytes both in vivo and in vitro. Mechanically, folic acid reduces the expression of m6A demethylase AlkB homolog 5 (ALKHB5) via promoter DNA hypermethylation. Decreased ALKBH5 causes increased m6A modification and increased expression of ATG12 in a demethylase activity-dependent manner, thereby promoting autophagy and preventing hepatic steatosis. Inhibition of ATG12 induced by overexpression of ALKBH5 could impair autophagy and the inhibitory effect of folic acid on lipid accumulation in hepatocytes. Together, these findings provide novel insights into understanding the protective role of folic acid in the treatment of NAFLD and suggest that folic acid may be a potential agent for combating NAFLD.
Collapse
Affiliation(s)
- Chaoqun Huang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Yaojun Luo
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Youhua Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Jiaqi Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Yushi Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Botao Zeng
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Xing Liao
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Yuxi Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Xinxia Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China.
| |
Collapse
|
4
|
Zhang L, Gan L, Lin Y, Mei Z, Liao S. FTO Promotes Hepatocellular Carcinoma Progression by Mediating m6A Modification of BUB1 and Targeting TGF-βR1 to Activate the TGF-β Signaling Pathway. J Clin Transl Hepatol 2025; 13:385-394. [PMID: 40385944 PMCID: PMC12078176 DOI: 10.14218/jcth.2025.00007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/01/2025] [Accepted: 03/17/2025] [Indexed: 05/20/2025] Open
Abstract
Background and Aims Fat mass and obesity-associated protein (FTO) has been linked to various cancers, though its role in hepatocellular carcinoma (HCC) remains unclear. This study aimed to investigate FTO expression, its clinical relevance, functional role in HCC progression, and the underlying molecular mechanisms. Methods Quantitative reverse-transcription polymerase chain reaction and immunohistochemical analysis were used to assess FTO expression in HCC. Functional assays, including proliferation, invasion, and epithelial-mesenchymal transition studies, were conducted using HCC cell lines with FTO knockdown. N6-methyladenosine (m6A) RNA immunoprecipitation and RNA stability assays further elucidated the role of FTO in BUB1 mRNA methylation and stability. Co-immunoprecipitation studies were employed to confirm the interaction between BUB1 and TGF-βR1. In vivo studies in nude mice were conducted to evaluate tumor growth following FTO knockdown. Results FTO was significantly upregulated in HCC tissues compared to normal liver tissues, with higher expression observed in advanced tumor-node-metastasis stages and metastatic HCC. Elevated FTO correlated with poor overall survival in patients. Silencing FTO decreased HCC cell proliferation, colony formation, invasion, epithelial-mesenchymal transition, and tumor growth in nude mice. Mechanistically, FTO downregulation led to increased m6A modification of BUB1 mRNA, thereby promoting its degradation via the YTH domain family 2-dependent pathway and reducing BUB1 protein levels. Additionally, BUB1 physically interacted with TGF-βR1, activating downstream TGF-β signaling. Conclusions FTO is overexpressed in HCC and is associated with poor clinical outcomes. Mechanistically, FTO promotes HCC progression by stabilizing BUB1 mRNA through an m6A-YTH domain family 2-dependent pathway, which activates TGF-β signaling. Targeting the FTO-BUB1-TGF-βR1 regulatory network may offer a promising therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Gastroenterology, Chongqing Jiangjin Central Hospital, Chongqing, China
| | - Li Gan
- Department of Anatomy, and Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, China
| | - Yuru Lin
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhechuan Mei
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shengtao Liao
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
5
|
Luo Y, Luo W, Cao Y, Wang Z. m6A demethylase FTO/ALKBH5 promotes diabetes-induced endothelial cell dysfunction by negatively regulating lncRNA H19. Exp Mol Pathol 2025; 143:104970. [PMID: 40381572 DOI: 10.1016/j.yexmp.2025.104970] [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: 10/09/2024] [Revised: 05/02/2025] [Accepted: 05/07/2025] [Indexed: 05/20/2025]
Abstract
Endothelial cell dysfunction induced by glucose is the most important cause of diabetic vascular complications, which are the leading causes of blindness, disability, renal failure, heart failure, stroke, and even death in diabetic patients. RNA m6A modification is involved in the pathogenesis of human disease. However, the role and underlying mechanism of RNA m6A modification in high glucose-induced endothelial cell dysfunction is not well understood. Herein, this study first demonstrated that m6A levels were decreased and that the demethylases FTO and ALKBH5 were upregulated in diabetic patients and an STZ-induced diabetic mouse model. This study revealed that high glucose induced decreased m6A levels and increased expression of FTO and ALKBH5, and silencing of FTO and ALKBH5 restored high glucose-induced decreases in m6A levels and dysfunction of HUVECs. Next, this study systematically screened differentially expressed lncRNAs, including H19, in HUVECs under high glucose conditions. This study revealed that FTO-ALKBH5 inhibited H19 expression by decreasing m6A modification in H19 transcripts. In addition, this study demonstrated the role of the FTO/ALKBH5/H19 pathway in high glucose-induced cellular dysfunction of HUVECs. Ultimately, this study uncovered that silencing of H19 promoted the expression of cell cycle-related genes, including PTEN, p21 and p27 via interacting with EZH2 and affecting the H3K27me3 histone modification. Overall, this study is the first to dissect the regulation of lncRNA by m6A modification in hyperglycaemia, identifying a new regulatory pathway in high glucose-induced cellular dysfunction and providing biomarkers with the potential to serve as therapeutic targets for high glucose-induced cellular dysfunction.
Collapse
Affiliation(s)
- Yanli Luo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China.
| | - Wanjun Luo
- Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Yanan Cao
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Zhanpeng Wang
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 410008, PR China
| |
Collapse
|
6
|
Davis CN, Khan Y, Toikumo S, Jinwala Z, Boomsma DI, Levey DF, Gelernter J, Kember RL, Kranzler HR. Integrating HiTOP and RDoC frameworks part II: shared and distinct biological mechanisms of externalizing and internalizing psychopathology. Psychol Med 2025; 55:e137. [PMID: 40340892 DOI: 10.1017/s0033291725000819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
BACKGROUND The Hierarchical Taxonomy of Psychopathology (HiTOP) and Research Domain Criteria (RDoC) frameworks emphasize transdiagnostic and mechanistic aspects of psychopathology. We used a multi-omics approach to examine how HiTOP's psychopathology spectra (externalizing [EXT], internalizing [INT], and shared EXT + INT) map onto RDoC's units of analysis. METHODS We conducted analyses across five RDoC units of analysis: genes, molecules, cells, circuits, and physiology. Using genome-wide association studies from the companion Part I article, we identified genes and tissue-specific expression patterns. We used drug repurposing analyses that integrate gene annotations to identify potential therapeutic targets and single-cell RNA sequencing data to implicate brain cell types. We then used magnetic resonance imaging data to examine brain regions and circuits associated with psychopathology. Finally, we tested causal relationships between each spectrum and physical health conditions. RESULTS Using five gene identification methods, EXT was associated with 1,759 genes, INT with 454 genes, and EXT + INT with 1,138 genes. Drug repurposing analyses identified potential therapeutic targets, including those that affect dopamine and serotonin pathways. Expression of EXT genes was enriched in GABAergic, cortical, and hippocampal neurons, while INT genes were more narrowly linked to GABAergic neurons. EXT + INT liability was associated with reduced gray matter volume in the amygdala and subcallosal cortex. INT genetic liability showed stronger causal effects on physical health - including chronic pain and cardiovascular diseases - than EXT. CONCLUSIONS Our findings revealed shared and distinct pathways underlying psychopathology. Integrating genomic insights with the RDoC and HiTOP frameworks advanced our understanding of mechanisms that underlie EXT and INT psychopathology.
Collapse
Affiliation(s)
- Christal N Davis
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA
- Department of Psychiatry, Center for Studies of Addiction, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Yousef Khan
- Department of Psychiatry, Center for Studies of Addiction, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Sylvanus Toikumo
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA
- Department of Psychiatry, Center for Studies of Addiction, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Zeal Jinwala
- Department of Psychiatry, Center for Studies of Addiction, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Dorret I Boomsma
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, The Netherlands and Amsterdam Reproduction and Development Research Institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Daniel F Levey
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Psychiatry Division, VA Connecticut Healthcare Center, West Haven, CT, USA
| | - Joel Gelernter
- Psychiatry Division, VA Connecticut Healthcare Center, West Haven, CT, USA
- Departments of Psychiatry, Genetics, and Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Rachel L Kember
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA
- Department of Psychiatry, Center for Studies of Addiction, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Henry R Kranzler
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA
- Department of Psychiatry, Center for Studies of Addiction, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
7
|
Melnik BC, Weiskirchen R, John SM, Stremmel W, Leitzmann C, Weiskirchen S, Schmitz G. White Adipocyte Stem Cell Expansion Through Infant Formula Feeding: New Insights into Epigenetic Programming Explaining the Early Protein Hypothesis of Obesity. Int J Mol Sci 2025; 26:4493. [PMID: 40429638 PMCID: PMC12110815 DOI: 10.3390/ijms26104493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 05/03/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Prolonged breastfeeding (BF), as opposed to artificial infant formula feeding (FF), has been shown to prevent the development of obesity later in life. The aim of our narrative review is to investigate the missing molecular link between postnatal protein overfeeding-often referred to as the "early protein hypothesis"-and the subsequent transcriptional and epigenetic changes that accelerate the expansion of adipocyte stem cells (ASCs) in the adipose vascular niche during postnatal white adipose tissue (WAT) development. To achieve this, we conducted a search on the Web of Science, Google Scholar, and PubMed databases from 2000 to 2025 and reviewed 750 papers. Our findings revealed that the overactivation of mechanistic target of rapamycin complex 1 (mTORC1) and S6 kinase 1 (S6K1), which inhibits wingless (Wnt) signaling due to protein overfeeding, serves as the primary pathway promoting ASC commitment and increasing preadipocyte numbers. Moreover, excessive protein intake, combined with the upregulation of the fat mass and obesity-associated gene (FTO) and a deficiency of breast milk-derived microRNAs from lactation, disrupts the proper regulation of FTO and Wnt pathway components. This disruption enhances ASC expansion in WAT while inhibiting brown adipose tissue development. While BF has been shown to have protective effects against obesity, the postnatal transcriptional and epigenetic changes induced by excessive protein intake from FF may predispose infants to early and excessive ASC commitment in WAT, thereby increasing the risk of obesity later in life.
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;
| | - 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
| | | | - Claus Leitzmann
- Institut für Ernährungswissenschaft, Universität Gießen, D-35392 Gießen, Germany;
| | - Sabine Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, D-52074 Aachen, Germany;
| | - Gerd Schmitz
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital of Regensburg, D-93053 Regensburg, Germany;
| |
Collapse
|
8
|
Shang M, Qin J, Zhao B, Luo Q, Wang H, Yang C, Liu H, Ran J, Yang W. Recent Advance in Sensitive Detection of Demethylase FTO. Chembiochem 2025; 26:e202400995. [PMID: 39714929 DOI: 10.1002/cbic.202400995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 12/24/2024]
Abstract
Methylation modification is a critical regulatory mechanism in epigenetics and plays a significant role in various biological processes. N6-methyladenosine (m6A) is the most common modification found in RNA. The fat mass and obesity-associated protein (FTO) facilitate the demethylation of m6A in RNA, and its abnormal expression is closely linked to the development of several diseases. As a result, FTO has the potential to serve as an important biomarker for clinical disease diagnosis. Despite its significance, there has been a lack of comprehensive reviews addressing advancements in detection methods for the demethylase FTO. This review provides an overview of the progress in FTO detection methods, ranging from traditional approaches to innovative techniques, with a particular emphasis on recently reported advancements. These novel detection methods can be categorized into strategies based on enzymes, functional nucleic acids (FNA), and conformational changes. We summarize the principles and applications of these detection methods and discuss the current challenges and prospects in this field.
Collapse
Affiliation(s)
- Min Shang
- College of Biological and Pharmaceutical Sciences, Key Laboratory of Natural Products, China Three Gorges University, 443002, Yichang, Hubei, P. R. China
| | - Jingying Qin
- College of Biological and Pharmaceutical Sciences, Key Laboratory of Natural Products, China Three Gorges University, 443002, Yichang, Hubei, P. R. China
| | - Bingyue Zhao
- College of Biological and Pharmaceutical Sciences, Key Laboratory of Natural Products, China Three Gorges University, 443002, Yichang, Hubei, P. R. China
| | - Qian Luo
- College of Biological and Pharmaceutical Sciences, Key Laboratory of Natural Products, China Three Gorges University, 443002, Yichang, Hubei, P. R. China
| | - Huimin Wang
- College of Biological and Pharmaceutical Sciences, Key Laboratory of Natural Products, China Three Gorges University, 443002, Yichang, Hubei, P. R. China
| | - Changying Yang
- College of Biological and Pharmaceutical Sciences, Key Laboratory of Natural Products, China Three Gorges University, 443002, Yichang, Hubei, P. R. China
| | - Hanghang Liu
- College of Biological and Pharmaceutical Sciences, Key Laboratory of Natural Products, China Three Gorges University, 443002, Yichang, Hubei, P. R. China
| | - Jiabing Ran
- College of Biological and Pharmaceutical Sciences, Key Laboratory of Natural Products, China Three Gorges University, 443002, Yichang, Hubei, P. R. China
| | - Wei Yang
- College of Biological and Pharmaceutical Sciences, Key Laboratory of Natural Products, China Three Gorges University, 443002, Yichang, Hubei, P. R. China
| |
Collapse
|
9
|
Jafari A, Khoshdooz S, Bafrani MA, Bakhshimoghaddam F, Abbasi H, Doaei S. Uncovering the Causal Link Between Obesity-Associated Genes and Multiple Sclerosis: A Systematic Literature Review. Brain Behav 2025; 15:e70439. [PMID: 40195065 PMCID: PMC11975544 DOI: 10.1002/brb3.70439] [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: 10/11/2024] [Revised: 02/25/2025] [Accepted: 03/02/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is a multifaceted neurodegenerative disorder influenced by genetics and lifestyle. This systematic literature review investigates the role of six obesity-associated genes, including fat mass and obesity-associated (FTO), FAS apoptosis inhibitory molecule 2 (FAIM2), Niemann-Pick disease type C1-like 1 (NPC1), glucosamine-6-phosphate deaminase 2 (GNPDA2), melanocortin-4 receptor (MC4R), and brain-derived neurotrophic factor (BDNF) in the context of MS. METHODS A literature search was executed using Embase, Scopus, Cochrane, Web of Science, and PubMed databases from inception to July 2024. The related keywords employed during the search process are "fas apoptotic inhibitory molecule 2," "Niemann-Pick disease type C1," "fat mass and obesity-associated," "melanocortin-4 receptor," "brain-derived neurotrophic factor," "glucosamine-6-phosphate deaminase 2," and "multiple sclerosis." RESULTS Out of 2108 papers, 27 were entered into the present systematic literature review. The FTO gene may affect MS susceptibility through metabolic and inflammatory pathways. FAIM2 and NPC1 genes may contribute to MS pathogenesis, though their precise roles are still being elucidated. The GNPDA2 gene may have some connections with MS but requires further clarification. MC4R has demonstrated significant neuroprotective and anti-inflammatory effects, suggesting its potential impact on MS progression. BDNF plays a complex role in neuronal survival and repair and may influence the risk of MS. CONCLUSION Our findings demonstrated that obesity-related genes may have a significant impact on MS risk and disease course, revealing novel insights into the genetic underpinnings of MS.
Collapse
Affiliation(s)
- Ali Jafari
- Student Research Committee, Department of Community Nutrition, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research InstituteShahid Beheshti University of Medical SciencesTehranIran
- Systematic Review and Meta‐analysis Expert Group (SRMEG)Universal Scientific Education and Research Network (USERN)TehranIran
| | - Sara Khoshdooz
- Faculty of MedicineGuilan University of Medical ScienceRashtIran
| | | | - Farnush Bakhshimoghaddam
- Department of Nutrition, School of Allied Medical SciencesAhvaz Jundishapur University of Medical SciencesAhvazIran
| | - Hamid Abbasi
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
- Neurosciences Research CenterTabriz University of Medical SciencesTabrizIran
| | - Saeid Doaei
- Department of Community Nutrition, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research InstituteShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
10
|
Fatima SS, Fatimi AS, Abbas M, Farhat S, Mohammed N. Methylation Patterns of Diabetes and Obesity Susceptibility Genes in Gestational Diabetes Mellitus: A Cross-Sectional Analysis from Karachi, Pakistan. Metab Syndr Relat Disord 2025; 23:137-145. [PMID: 39515368 DOI: 10.1089/met.2024.0171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Background: Women with gestational diabetes mellitus (GDM) and their offspring have an increased risk of adverse perinatal and long-term health outcomes, which may be attributable to epigenetic modification of diabetes and obesity susceptibility genes. We aimed to investigate the methylation patterns of eight genes in GDM and normoglycemic (NG) mothers, and their respective offspring. Methods: This cross-sectional study, conducted at Aga Khan University from August 2019 to December 2022, recruited pregnant women in the first trimester of gestation from the outpatient obstetrics clinic. Participants were classified as NG or GDM based on the Society of Obstetricians and Gynecologists Pakistan. Venous blood samples were collected from mothers and cord blood from neonates. Peripheral blood mononuclear cells were used for DNA extraction and methylation analysis using methylation-specific PCR. Maternal and neonatal clinical data were recorded. Statistical analysis was performed using R, including binary logistic regression to assess the association between various gene methylation levels and GDM. Results: The study found that GDM mothers had significantly higher fasting blood glucose, 2-hr OGTT, and serum carboxymethyl lysine (CML) levels compared to NG mothers, but no significant differences in neonatal birth weight or serum CML levels. Chemerin methylation was significantly lower in GDM mothers and their babies, while NAMPT, MTNR1B, FNDC5, FAT4, and FTO methylation levels were higher in GDM offspring compared to NG offspring. GDM mothers also had higher methylation levels of brain-derived neurotrophic factor gene (BDNF). Multivariable binary logistic regression identified methylation levels of maternal BDNF and neonatal MTNR1B to be independently associated with GDM. Conclusions: Our study shows a trend of epigenetic modifications in both GDM mothers and their offspring in various genes related to metabolism and inflammation, suggesting an intergenerational transmission of increased risk of developing metabolic disorders. These findings emphasize the need for high throughput studies, early screening, tight glucose control during pregnancy, and postnatal follow-up to mitigate long-term health risks.
Collapse
Affiliation(s)
- Syeda Sadia Fatima
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | | | - Manzar Abbas
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | - Sabah Farhat
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | - Nuruddin Mohammed
- Department of Obstetrics and Gynecology, Aga Khan University, Karachi, Pakistan
| |
Collapse
|
11
|
Bazzazzadehgan S, Shariat-Madar Z, Mahdi F. Distinct Roles of Common Genetic Variants and Their Contributions to Diabetes: MODY and Uncontrolled T2DM. Biomolecules 2025; 15:414. [PMID: 40149950 PMCID: PMC11940602 DOI: 10.3390/biom15030414] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/26/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) encompasses a range of clinical manifestations, with uncontrolled diabetes leading to progressive or irreversible damage to various organs. Numerous genes associated with monogenic diabetes, exhibiting classical patterns of inheritance (autosomal dominant or recessive), have been identified. Additionally, genes involved in complex diabetes, which interact with environmental factors to trigger the disease, have also been discovered. These genetic findings have raised hopes that genetic testing could enhance diagnostics, disease surveillance, treatment selection, and family counseling. However, the accurate interpretation of genetic data remains a significant challenge, as variants may not always be definitively classified as either benign or pathogenic. Research to date, however, indicates that periodic reevaluation of genetic variants in diabetes has led to more consistent findings, with biases being steadily eliminated. This has improved the interpretation of variants across diverse ethnicities. Clinical studies suggest that genetic risk information may motivate patients to adopt behaviors that promote the prevention or management of T2DM. Given that the clinical features of certain monogenic diabetes types overlap with T2DM, and considering the significant role of genetic variants in diabetes, healthcare providers caring for prediabetic patients should consider genetic testing as part of the diagnostic process. This review summarizes current knowledge of the most common genetic variants associated with T2DM, explores novel therapeutic targets, and discusses recent advancements in the pharmaceutical management of uncontrolled T2DM.
Collapse
Affiliation(s)
- Shadi Bazzazzadehgan
- Department of Pharmacy Administration, School of Pharmacy, University of Mississippi, University, MS 38677, USA;
| | - Zia Shariat-Madar
- Division of Pharmacology, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA;
| | - Fakhri Mahdi
- Division of Pharmacology, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA;
| |
Collapse
|
12
|
Irisarri A, Corral A, Perez-Salvador N, Bellver-Sanchis A, Ribalta-Vilella M, Bentanachs R, Alegret M, Laguna JC, Barroso E, Palomer X, Ortuño-Sahagún D, Vázquez-Carrera M, Pallàs M, Herrero L, Griñán-Ferré C. FTO inhibition mitigates high-fat diet-induced metabolic disturbances and cognitive decline in SAMP8 mice. Mol Med 2025; 31:73. [PMID: 39984825 PMCID: PMC11843768 DOI: 10.1186/s10020-025-01126-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/12/2025] [Indexed: 02/23/2025] Open
Abstract
This study investigated the effects of fat mass and obesity-associated (FTO) inhibition on cognitive function and metabolic parameters of senescence-accelerated mouse prone 8 (SAMP8) mice fed a high-fat diet (HFD). SAMP8 mice fed an HFD exhibited increased body weight, impaired glucose tolerance, and elevated serum leptin levels. In epididymal white adipose tissue (eWAT), pharmacological treatment with FB23, a well-established FTO inhibitor, increased leptin production and modulated genes involved in lipid metabolism (Cpt1a, Atgl, Hsl, Fas), oxidative stress (OS) (Bip, Edem), and inflammation (Mcp1, Tnfα). Expression of hepatic genes related to lipid metabolism (Cpt1a, Atgl, Mgl, Dgat2, Srebp, Plin2) and OS (catalase, Edem) were modulated by FB23, although hepatic steatosis remained unchanged. Remarkably, FB23 treatment increased m6A RNA methylation in the brain, accompanied by changes in N6-methyladenosine (m6A)-regulatory enzymes and modulation of neuroinflammatory markers (Il6, Mcp1, iNOS). FTO inhibition reduced the activity of matrix metalloproteases (Mmp2, Mmp9) and altered IGF1 signaling (Igf1, Pten). Notably, enhanced leptin signaling was observed through increased expression of immediate early genes (Arc, Fos) and the transcription factor Stat3. Improved synaptic plasticity was evident, as shown by increased levels of neurotrophic factors (Bdnf, Ngf) and restored neurite length and spine density. Consistent with these findings, behavioral tests demonstrated that FB23 treatment effectively rescued cognitive impairments in SAMP8 HFD mice. The novel object recognition test (NORT) and object location test (OLT) revealed that treated mice exhibited enhanced short- and long-term memory and spatial memory compared to the HFD control group. Additionally, the open field test showed a reduction in anxiety-like behavior after treatment with FB23. In conclusion, pharmacological FTO inhibition ameliorated HFD-induced metabolic disturbances and cognitive decline in SAMP8 mice. These results suggest that targeting FTO may be a promising therapeutic approach to counteract obesity-induced cognitive impairment and age-related neurodegeneration.
Collapse
Grants
- PID2021-122116OB-100 Ministerio de Economía, Industria Economía, Industria y Competitividad (Agencia Estatal de Investigación, AEI) and European Union NextGenerationEU/PRTR
- PDC2022-133441-I00 Ministerio de Economía, Industria Economía, Industria y Competitividad (Agencia Estatal de Investigación, AEI) and European Union NextGenerationEU/PRTR
- PID2020-114953RB-C21 Ministerio de Economía, Industria Economía, Industria y Competitividad (Agencia Estatal de Investigación, AEI) and European Union NextGenerationEU/PRTR
- PID2022-139016OA-I00 Ministerio de Economía, Industria Economía, Industria y Competitividad (Agencia Estatal de Investigación, AEI) and European Union NextGenerationEU/PRTR
- CIBERDEM CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)
- 2021 SGR 00357 the Government of Catalonia
- CB06/03/0001 CIBEROBN
- 2021SGR00367 Carlos III Health Institute project and the Government of Catalonia
- Producte 0092 Departament d'Empresa i Coneixement de la Generalitat de Catalunya 2023
- Llavor 005 and 007 Departament d'Empresa i Coneixement de la Generalitat de Catalunya 2023
- Departament d’Empresa i Coneixement de la Generalitat de Catalunya 2023
Collapse
Affiliation(s)
- Alba Irisarri
- pHD Program in Biotechnology, Faculty of Pharmacy and Food Sciences, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Institute of Neurosciences of the University of Barcelona, University of Barcelona, 08035, Barcelona, Spain
| | - Ana Corral
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
| | - Núria Perez-Salvador
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
| | - Aina Bellver-Sanchis
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Institute of Neurosciences of the University of Barcelona, University of Barcelona, 08035, Barcelona, Spain
| | - Marta Ribalta-Vilella
- pHD Program in Biotechnology, Faculty of Pharmacy and Food Sciences, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Institute of Neurosciences of the University of Barcelona, University of Barcelona, 08035, Barcelona, Spain
| | - Roger Bentanachs
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
| | - Marta Alegret
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Juan Carlos Laguna
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Emma Barroso
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM) - National Institute of Health Carlos III, 28029, Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, 08950, Esplugues de Llobregat, Barcelona, Spain
| | - Xavier Palomer
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM) - National Institute of Health Carlos III, 28029, Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, 08950, Esplugues de Llobregat, Barcelona, Spain
| | - Daniel Ortuño-Sahagún
- Laboratorio de Neuroinmunología Molecular, Instituto de Investigación de Ciencias Biomédicas (IICB) CUCS, Universidad de Guadalajara, 44340, Guadalajara, Jalisco, Mexico
| | - Manuel Vázquez-Carrera
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM) - National Institute of Health Carlos III, 28029, Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, 08950, Esplugues de Llobregat, Barcelona, Spain
| | - Mercè Pallàs
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Institute of Neurosciences of the University of Barcelona, University of Barcelona, 08035, Barcelona, Spain
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Herrero
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Christian Griñán-Ferré
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain.
- Institute of Neurosciences of the University of Barcelona, University of Barcelona, 08035, Barcelona, Spain.
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
13
|
Manzoori S, Torshizi RV, Masoudi AA, Momen M. Novel Candidate Genes Detection Using Bayesian Network-Based Genome-Wide Association Study of Latent Traits in F2 Chicken Population. J Anim Breed Genet 2025. [PMID: 39968732 DOI: 10.1111/jbg.12926] [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: 06/02/2024] [Revised: 12/01/2024] [Accepted: 01/16/2025] [Indexed: 02/20/2025]
Abstract
In chickens, economically important traits are commonly controlled by multiple genes and are often correlated. The genetic mechanisms underlying the correlated phenotypes likely involve pleiotropy or linkage disequilibrium, which is not handled properly in single-trait genome-wide association studies (GWAS). We employed factor analytical models to estimate the value of latent traits to reduce the dimensionality of the adjusted phenotypes. The dataset included phenotypes from 369 F2 chickens, categorised into six observable classes, namely body weight (BW), feed intake (FI), feed efficiency (FE), immunity (IMU), blood metabolites (BMB), and carcass (CC) traits. All birds were genotyped using a 60K SNP Beadchip. A Bayesian network (BN) algorithm was used to discern the recursive causal relationships among the inferred latent traits. Multi-Trait (MT) and Structural Equation Model (SEM) were applied for association analysis. Several candidate genes were detected across six phenotypic classes, namely the IPMK gene for BW and FI, and, the MTERF2 gene for BW and FE. The rs14565514 SNP, close to genes IPMK, UBE2D1, and CISD1, was recognised as a pleiotropic marker by both models. The NRG3 gene, located on chromosome 6, was associated with FI. CRISP2, RHAG, CYP2AC1, and CENPQ genes, located on chromosome 3, were detected for BMB through both MT- and SEM-GWAS. In general, the results indicated that the SEM-GWAS is superior to MT-GWAS due to considering the causal relationships among the traits, correcting the effects of the traits on each other, and also leading to the identification of pleiotropic SNP markers.
Collapse
Affiliation(s)
- Siavash Manzoori
- Department of Animal Science, Tarbiat Modares University, Tehran, Iran
| | | | - Ali Akbar Masoudi
- Department of Animal Science, Tarbiat Modares University, Tehran, Iran
| | - Mehdi Momen
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
14
|
Emmanuel OP, Sandrine MNY, Claude BD, Ronald BAG, Vicky AM, Désiré DDP, Pierre K, Aziz T, Alamri AS, Alsanie WF, Alhomrani M. Exploring the Effects of Pterocarpus Soyauxii Against Menopause-Related NAFLD Based on Network Pharmacology, Molecular Docking, and Experimental Validation. Chem Biodivers 2025:e202403384. [PMID: 39964816 DOI: 10.1002/cbdv.202403384] [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: 12/19/2024] [Revised: 02/12/2025] [Accepted: 02/18/2025] [Indexed: 02/20/2025]
Abstract
Pterocarpus soyauxii (P. soyauxii) is a Fabaceae family traditionally used to treat menopausal disorders. This study aims to investigate the effect of P. soyauxii on menopause-related non-alcoholic fatty liver disease (NAFLD) and to determine its mechanisms of action and signaling pathways. The pharmacokinetic and dynamic properties and the toxicological profile of P. soyauxii compounds were assessed using the SwissADME and Protox III databases. A pharmacology network was constructed to identify active compound targets and corresponding genes. Compound target and protein-protein interaction networks were created using Cytoscape software. Molecular docking studies were conducted to assess the binding affinity of P. soyauxii compounds with specific proteins. In vitro experiments evaluated the antioxidant properties of P. soyauxii. In vivo studies using ovariectomized (Ovx) rat models underlined pathways and effects of P. soyauxii on biochemical and histological features linked with NAFLD. Findings suggest that P. soyauxii compounds are readily absorbed through the intestine and exhibit a relatively low level of toxicity. Protein-protein interaction, compound-target networks, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed several pathways and target proteins of P. soyauxii compounds. Indeed, they target specific proteins such as estrogen receptor alpha/beta ERα/β, nicotinamide adenine dinucleotide phosphate oxidase (NADPH-O), epidermal growth factor receptor (EGFR), MAPK1, peroxisome proliferator-activated receptors alpha/gamma (PPARα/G), and HMG-CoA reductase. Molecular docking revealed that P. soyauxii compounds demonstrate high binding affinity to various proteins. In vitro, P. soyauxii inhibits the oxidative power of OH, H2O2, and NO. In vivo, P. soyauxii significantly (p < 0.01, p < 0.001, and p < 0.01, respectively) reduces ALAT (25.14%), hepatic cholesterol (15.27%), and malondialdehyde (MDA) (26.78%) levels at 200 mg/kg and prevents steatosis in the liver. These findings suggest that P. soyauxii may have a protective role against menopause-related NAFLD.
Collapse
Affiliation(s)
- Owona Pascal Emmanuel
- Department of Animal Biology and Physiology Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé 1, Yaoundé, Cameroon
| | - Mengue Ngadena Yolande Sandrine
- Department of Animal Biology and Physiology Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé 1, Yaoundé, Cameroon
- Neurosciences and Psychogerontology Axis, Laboratory of Development and Maldevelopment, Department of Psychology, Faculty of Arts, Letters, and Social Science, University of Yaoundé 1, Yaoundé, Cameroon
| | - Bilanda Danielle Claude
- Department of Animal Biology and Physiology Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé 1, Yaoundé, Cameroon
| | - Bidingha A Goufani Ronald
- Department of Animal Biology and Physiology Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé 1, Yaoundé, Cameroon
| | - Ama Moor Vicky
- Faculty of Medicine and Biomedical Sciences, Clinical Biochemistry Laboratory, University Hospital Centre, Yaoundé, Cameroon
| | - Dzeufiet Djomeni Paul Désiré
- Department of Animal Biology and Physiology Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé 1, Yaoundé, Cameroon
| | - Kamtchouing Pierre
- Department of Animal Biology and Physiology Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé 1, Yaoundé, Cameroon
| | - Tariq Aziz
- Laboratory of Animal Health Food Hygiene and Quality, University of Ioannina, Arta, Greece
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Abdulhakeem S Alamri
- Department of Clinical Laboratory Sciences, The faculty of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Walaa F Alsanie
- Department of Clinical Laboratory Sciences, The faculty of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Majid Alhomrani
- Department of Clinical Laboratory Sciences, The faculty of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| |
Collapse
|
15
|
Somala CS, Sathyapriya S, Bharathkumar N, Anand T, Mathangi DC, Saravanan KM. Therapeutic Potential of FTO Demethylase in Metabolism and Disease Pathways. Protein J 2025; 44:21-34. [PMID: 39923206 DOI: 10.1007/s10930-025-10250-3] [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] [Accepted: 01/25/2025] [Indexed: 02/10/2025]
Abstract
The crucial involvement of the Fat Mass and Obesity-associated (FTO) protein in both metabolic and non-metabolic diseases has been documented since its discovery. This enzyme, known as FTO, is a demethylase that belongs to the 2-oxoglutarate-dependent nucleic acid demethylases. Its primary function is to target N6-methyladenosine (m6A) in RNA, which is crucial in regulating RNA stability, processing, and expression. This review facilitates understanding the FTO gene variations linked to Body Mass Index (BMI) and obesity, resulting in increased vulnerability to type 2 diabetes. While prior reviews have already discussed the link between FTO and BMI and its impact on type 2 diabetes, the current review additionally examines the emerging evidence suggesting a direct influence of the FTO gene on metabolism. Additionally, the paper discusses the alternative role of FTO and emphasizes the endophenotypes in neurological circuits and the demethylase function of FTO in neurodegenerative disorders. The review further examines the impact of FTO on several physiological systems and emphasizes the need to study FTO as a potential multitarget for future research and therapies.
Collapse
Affiliation(s)
- Chaitanya Sree Somala
- Department of Mind Body Medicine and Lifestyle Sciences, Faculty of Allied Health Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, 600116, India
| | - Selvaraj Sathyapriya
- Sri Ramachandra Innovation Incubation Center (SRIIC) Lab, Faculty of Clinical Research, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, 600116, India
| | | | - Thirunavukarasou Anand
- Sri Ramachandra Innovation Incubation Center (SRIIC) Lab, Faculty of Clinical Research, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, 600116, India
| | - Damal Chandrasekar Mathangi
- Department of Mind Body Medicine and Lifestyle Sciences, Faculty of Allied Health Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, 600116, India.
| | - Konda Mani Saravanan
- B Aatral Biosciences Private Limited, Bangalore, Karnataka, 560091, India.
- Department of Biotechnology, Bharath Institute of Higher Education and Research, Chennai, Tamil Nadu, 600073, India.
| |
Collapse
|
16
|
Jamali MA, Abdeen SM, Mathew TC. Association of Fat Mass and Obesity (FTO) rs9939609 Single Nucleotide Polymorphism (SNP) With Obesity and Type 2 Diabetes (T2D) in Healthy Young Adults in Kuwait. Cureus 2025; 17:e77110. [PMID: 39925495 PMCID: PMC11802477 DOI: 10.7759/cureus.77110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2025] [Indexed: 02/11/2025] Open
Abstract
OBJECTIVE The aim was to determine the association of the common fat mass and obesity-associated (FTO) gene polymorphism rs9939609 with the risk of developing obesity and type 2 diabetes (T2D) in healthy young university students in Kuwait. METHODS This cross-sectional study included 201 students from Kuwait University (males = 99; females = 102). The author analyzed the association of FTO and obesity using the body mass index (BMI) as a binary categorical variable (non-obese-BMI < 25 vs. obese-BMI > 25) and the BMI as a continuous variable, using both logistic and linear regression models, respectively. Genotyping of the FTO rs9969609 was performed using the TaqMan single nucleotide polymorphism (SNP) Genotyping Assay (Applied Biosystems, Foster City, USA) and ABI 7500 Fast Real-Time PCR system SDS software (Life Technologies, California, USA) was used for allelic discrimination. RESULTS BMI (continuous variable) was not associated with FTO in either the adjusted dominant genetic model (p = 0.33) or the additive model (p = 0.35). Similarly, neither the adjusted dominant (p = 0.66) nor the additive model (p = 0.39) showed FTO as a significant predictor of categorical BMI. In addition, the analysis showed that FTO was not a risk factor for T2D in either the adjusted dominant (p = 0.08) or the additive model (p = 0.17). The FTO allele frequency (%) in the study population (A: 44%; T: 56%) did not significantly differ from the global allele frequency (p-value > 0.05). As for genotype frequency distribution (%) in this study, FTO (AG: 37.8%; AA: 5.5%; GG: 56.7%) was found not to be in equilibrium with the Hardy-Weinberg Equation. CONCLUSION FTO is not a significant predictor of obesity or the future risk of T2D in healthy young adults of Kuwait. The cross-sectional genetic study design is a limitation for the detection or significant predictors of obesity or T2D among young adults in Kuwait.
Collapse
Affiliation(s)
- Mohammed A Jamali
- Department of Psychiatry, Faculty of Medicine, Health Sciences Center, Kuwait University, Kuwait City, KWT
- Department of Pathology, Faculty of Medicine, Health Sciences Center, Kuwait University, Kuwait City, KWT
| | - Suad M Abdeen
- Department of Pathology, Faculty of Medicine, Health Sciences Center, Kuwait University, Kuwait City, KWT
| | - Thazhumpal C Mathew
- Department of Medical Laboratory Sciences, Faculty of Allied Health Science, Kuwait University, Kuwait City, KWT
| |
Collapse
|
17
|
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
|
18
|
Mandujano-Lázaro G, Torres-Rojas MF, Ramírez-Moreno E, Marchat LA. Virtual screening combined with molecular docking for the !identification of new anti-adipogenic compounds. Sci Prog 2025; 108:368504251320313. [PMID: 39936374 PMCID: PMC11815789 DOI: 10.1177/00368504251320313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
Obesity is an important risk factor for diabetes, cardiovascular diseases, and cancer, reducing the quality of life and expectancy of millions of people. Consequently, obesity has turned into one of the most health public problems worldwide, which highlights the urgent need for new and safe treatments. Obesity is mainly related to excessive fat accumulation; therefore, proteins participating in white adipose tissue increase and dysfunction are considered pertinent and attractive targets for developing new methods that can help with body weight control. In this context, virtual screening of libraries containing a large number of molecules represents a valuable strategy for the identification of potential anti-adipogenic compounds with reduced costs and time production. Here, we review the scientific literature about the prediction of new ligands of specific proteins through molecular docking and virtual screening of chemical libraries, with the aim of proposing new potential anti-adipogenic molecules. First, we present the targets related to adipogenesis and adipocyte functions that were selected for the following studies: PPARγ, Crif1, SIRT1, ERβ, PC1, FTO, Mss51, and FABP4. Then, we describe the obtention of new ligands according to the characteristics of the virtual screening approach, i.e. a structure-based drug design (SBDD) or a ligand-based drug design (LBDD). Finally, the critical analysis of these computational strategies and the corresponding results points out the necessity of combining computational and in vitro or in vivo assays for the identification of effective new anti-adipogenic molecules for obesity control. It also evidences that translating molecular docking and virtual screening results into successful drug candidates for adipogenesis and obesity control remains a huge challenge.
Collapse
Affiliation(s)
- Gilberto Mandujano-Lázaro
- Laboratorio de Biomedicina Molecular 2, ENMH, Instituto Politécnico Nacional, Ciudad de México, México
| | - María F Torres-Rojas
- Laboratorio de Biomedicina Molecular 2, ENMH, Instituto Politécnico Nacional, Ciudad de México, México
| | - Esther Ramírez-Moreno
- Laboratorio de Biomedicina Molecular 2, ENMH, Instituto Politécnico Nacional, Ciudad de México, México
| | - Laurence A Marchat
- Laboratorio de Biomedicina Molecular 2, ENMH, Instituto Politécnico Nacional, Ciudad de México, México
| |
Collapse
|
19
|
Banerjee M, Pandey VP. Diet-induced Obesity: Pathophysiology, Consequences and Target Specific Therapeutic Strategies. Curr Protein Pept Sci 2025; 26:113-124. [PMID: 39225225 DOI: 10.2174/0113892037329528240827180820] [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: 05/09/2024] [Revised: 07/09/2024] [Accepted: 07/18/2024] [Indexed: 09/04/2024]
Abstract
Diet has emerged as a pivotal factor in the current time for diet-induced obesity (DIO). A diet overloaded with fats and carbohydrates and unhealthy dietary habits contribute to the development of DIO through several mechanisms. The prominent ones include the transition of normal gut microbiota to obese microbiota, under-expression of AMPK, and abnormally high levels of adipogenesis. DIO is the root of many diseases. The present review deals with various aspects of DIO and its target proteins that can be specifically used for its treatment. Also, the currently available treatment strategies have been explored. It was found that the expression of five proteins, namely, PPARγ, FTO, CDK4, 14-3-3 ζ protein, and Galectin-1, is upregulated in DIO. They can be used as potential targets for drug-designing studies. Thus, with these targets, the treatment strategy for DIO using natural bioactive compounds can be a safer alternative to medications and bariatric surgeries.
Collapse
Affiliation(s)
- Munmun Banerjee
- Department of Biochemistry, University of Lucknow, Lucknow 226007, India
| | - Veda P Pandey
- Department of Biochemistry, University of Lucknow, Lucknow 226007, India
- Institute of Food Processing and Technology, ONGC Centre for Advanced Studies, University of Lucknow, Lucknow 226007, India (Pesent Address)
| |
Collapse
|
20
|
Parveen S, Khan S, Moin S, Nigar A, Khan MM, Alam R. Association of FTO protein with hyperandrogenism metabolic disturbances in women with polycystic ovary syndrome. Endocr Regul 2025; 59:33-41. [PMID: 40258223 DOI: 10.2478/enr-2025-0005] [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] [Indexed: 04/23/2025] Open
Abstract
Objective. Polycystic ovary syndrome (PCOS) is one of the commonest endocrinopathies in women characterized by hyperandrogenism, ovulatory dysfunction, and insulin resistance affecting 5-20% of reproductive-aged women worldwide. Recent studies have emphasized the role of the fat mass and obesity-associated (FTO) gene in the development of PCOS, specifically the rs9939609 A/T polymorphism, which is linked to an increased risk of PCOS. The study aimed to investigate the levels of FTO protein and its association with luteinizing hormone (LH), follicle-stimulating hormone (FSH) and anthropometric parameters in patients with PCOS compared to healthy controls. Materials. A total of 298 women, comprising 149 patients and 149 healthy controls, enrolled in the study. Anthropometric parameters (body mass index, BMI; waist circumference, WC; hip circumference, HC; waist-to-hip ratio, WHR), and hormonal assays (LH, FSH, LH/FSH ratio) were performed. FTO protein levels were measured by ELISA kit and their association with these parameters was analyzed. A receiver operator characteristic (ROC) curve analysis was performed to evaluate the discriminatory power of FTO protein levels in distinguishing PCOS cases and controls. A value p<0.05 was considered statistically significant. Results. FTO protein levels were significantly elevated in PCOS women with increased BMI, WC, HC, and WHR (p=<0.05). The mean of BMI showed a positive correlation with both WC (r=0.367, p<0.001) and HC (r=0.395, p<0.001). WC strongly correlated with HC (r=0.780, p<0.001) and WHR (r=0.465, p<0.001). LH significantly correlated with FSH (r=0.543, p<.001), and LH/FSH (r=0.553, p<.001). FTO protein showed a positive correlation with LH (r=0.364, p<0.001), and FSH (r=0.166, p<0.001). Additionally, a negative correlation of FTO protein with BMI (r=-0.190, p<0.05), WC (r=-0.277, p<0.05), and WHR (r=-0.408 p<0.001) was observed. The levels of FTO protein were significantly higher in PCOS patients compared to controls. Significant correlations were also found between FTO protein levels and the anthropometric or hormonal parameters. The AUC for FTO protein levels was 0.624 (p=0.550), indicating moderate discriminatory power, but lacking statistical significance. Conclusion. The study found that FTO protein levels are significantly higher in PCOS women correlating with anthropometric and hormonal parameters (increased LH, decreased FSH). This highlights potential involvement of FTO protein in the hormonal and metabolic disturbances characteristics of the syndrome indicating its biomarker character for the condition.
Collapse
Affiliation(s)
- Sadaf Parveen
- 1Department of Biochemistry, Integral Institute of Medical Sciences & Research (IIMSR), Integral University, Lucknow, Uttar Pradesh, India
| | - Saba Khan
- 1Department of Biochemistry, Integral Institute of Medical Sciences & Research (IIMSR), Integral University, Lucknow, Uttar Pradesh, India
| | - Shagufta Moin
- 2Department of Biochemistry, Jawaharlal Nehru Medical College Aligarh Muslim University Aligarh, India
| | - Asma Nigar
- 3Department of Obstetrics and Gynaecology, Integral Institute of Medical Sciences & Research (IIMSR), Integral University, Lucknow, Uttar Pradesh, India
| | - Mohammad Mustufa Khan
- 4Department of Basic Medical Sciences, Integral Institute of Allied Health Sciences & Research (IIAHSR), Integral University, Lucknow, Uttar Pradesh, India
| | - Roshan Alam
- 1Department of Biochemistry, Integral Institute of Medical Sciences & Research (IIMSR), Integral University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
21
|
Ahamba IS, Mary-Cynthia Ikele C, Kimpe L, Goswami N, Wang H, Li Z, Ren Z, Dong X. Unraveling the genetic and epigenetic landscape governing intramuscular fat deposition in rabbits: Insights and implications. FOOD CHEMISTRY. MOLECULAR SCIENCES 2024; 9:100222. [PMID: 39290671 PMCID: PMC11406001 DOI: 10.1016/j.fochms.2024.100222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/23/2024] [Accepted: 08/25/2024] [Indexed: 09/19/2024]
Abstract
Intramuscular fat (IMF) content is a predominant factor recognized to affect rabbit meat quality, directly impacting flavor, juiciness, and consumer preference. Despite its significance, the major interplay of genetic and epigenetic factors regulating IMF in rabbits remains largely unexplored. This review sheds light on this critical knowledge gap, offering valuable insights and future directions. We delve into the potential role of established candidate genes from other livestock (e.g. PPARγ, FABP4, and SCD) in rabbits, while exploring the identified novel genes of IMF in rabbits. Furthermore, we explored the quantitative trait loci studies in rabbit IMF and genomic selection approaches for improving IMF content in rabbits. Beyond genetics, this review unveils the exciting realm of epigenetic mechanisms modulating IMF deposition. We explored the potential of DNA methylation patterns, histone modifications, and non-coding RNA-mediation as fingerprints for selecting rabbits with desirable IMF levels. Additionally, we explored the possibility of manipulating the epigenetic landscape through nutraceuticals interventions to promote favorable IMF depositions. By comprehensively deciphering the genomic and epigenetic terrain of rabbit intramuscular fat regulation, this study aims to assess the existing knowledge regarding the genetic and epigenetic factors that control the deposition of intramuscular fat in rabbits. By doing so, we identified gaps in the current research, and suggested potential areas for further investigation that would enhance the quality of rabbit meat. This can enable breeders to develop targeted breeding strategies, optimize nutrition, and create innovative interventions to enhance the quality of rabbit meat, meet consumer demands and increase market competitiveness.
Collapse
Affiliation(s)
- Ifeanyi Solomon Ahamba
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, China
| | | | - Lionel Kimpe
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, China
| | - Naqash Goswami
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, China
| | - Hui Wang
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, China
| | - Zhen Li
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, China
| | - Zhanjun Ren
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, China
| | - Xianggui Dong
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, China
| |
Collapse
|
22
|
Obara-Gołębiowska M, Przybyłowicz KE, Danielewicz A, Sawicki T. Body mass as a result of psychological, lifestyle and genetic determinants. A pilot study involving overweight/obese and normal weight women in their early adulthood. PLoS One 2024; 19:e0314942. [PMID: 39671348 PMCID: PMC11642964 DOI: 10.1371/journal.pone.0314942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 11/19/2024] [Indexed: 12/15/2024] Open
Abstract
AIM The causes of obesity and overweight are complex and depend on mutually interrelated groups of lifestyle, psychological and genetic factors. Among 46 identified point mutations known within FTO gene, mutation SNP rs9939609 has the strongest effect on an increase in body weight. Therefore, the study aimed to assess psychological, lifestyle and genetic factors (expressed by the frequency of the FTO SNP rs-9939609 gene variant) and their association with body weight in young adult women. METHODS We genotyped FTO rs9939609 SNP in cheek swabs collected from 49 women aged 18-35, equally with and without overweight and obesity. Eating behaviour was defined based on the Questionnaire of Eating-Related Behaviors (QERB) and physical activity by the International Physical Activity Questionnaire (IPAQ). Classical anthropometric indices and the body composition analysis results measured by bioelectrical impedance were used to characterise nutritional status. RESULTS Overweight/obese participants had significantly higher body composition parameters than normal-weight participants, along with lower physical activity levels and more time spent sitting. Overweight/obese women presented significantly higher scores in emotional overeating, habitual overeating, and dietary restrictions, indicating more problematic eating behaviors compared to normal-weight individuals. No significant differences were observed in BMI, lifestyle factors, or eating behaviors across FTO gene variants (AA, AT, and TT). However, the TT and AT FTO gene variant groups showed significant positive correlations between habitual overeating and key measures of body weight (BMI, WC, and FM). In contrast, the AA variant group exhibited fewer associations between psychological and lifestyle factors with body weight. CONCLUSIONS Psychological and lifestyle factors, particularly overeating behaviors, were more strongly associated with increased body weight in women with the TT and AT variants of the FTO SNP rs-9939609 gene, highlighting the potential influence of genetic predisposition on eating habits and weight status in this population. Proper eating habits and high physical activity play an important role in preventing overweight and obesity regardless of the genotype that poses a potential risk of weight gain. The study's findings bring practical implications for health education and health psychology.
Collapse
Affiliation(s)
- Małgorzata Obara-Gołębiowska
- Faculty of Social Sciences, Department of Clinical, Developmental and Educational Psychology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | | | - Anna Danielewicz
- Faculty of Food Sciences, Department of Human Nutrition, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Tomasz Sawicki
- Faculty of Food Sciences, Department of Human Nutrition, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| |
Collapse
|
23
|
He YZ, Li XN, Li HT, Bai XH, Liu YC, Li FN, Lv BL, Qi TJ, Zhao XM, Li S. FTO promotes gefitinib-resistance by enhancing PELI3 expression and autophagy in non-small cell lung cancer. Pulm Pharmacol Ther 2024; 87:102317. [PMID: 39154901 DOI: 10.1016/j.pupt.2024.102317] [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: 05/22/2024] [Revised: 07/27/2024] [Accepted: 08/11/2024] [Indexed: 08/20/2024]
Abstract
The established recognition of N6-methyladenosine (m6A) modification as an indispensable regulatory agent in human cancer is widely accepted. However, the understanding of m6A's role and the mechanisms underlying its contribution to gefitinib resistance is notably limited. Herein, using RT-qPCR, Western blot, Cell proliferation and apoptosis, as well as RNA m6A modification assays, we substantiated that heightened FTO (Fat Mass and Obesity-associated protein) expression substantially underpins the emergence of gefitinib resistance in NSCLC cells. This FTO-driven gefitinib resistance is hinged upon the co-occurrence of PELI3 (Pellino E3 Ubiquitin Protein Ligase Family Member 3) expression and concurrent autophagy activation. Manipulation of PELI3 expression and autophagy activation, including its attenuation, was efficacious in both inducing and overcoming gefitinib resistance within NSCLC cells, as validated in vitro and in vivo. In summary, this study has successfully elucidated the intricate interplay involving FTO-mediated m6A modification, its consequential downstream effect on PELI3, and the concurrent involvement of autophagy in fostering the emergence of gefitinib resistance within the therapeutic context of NSCLC.
Collapse
Affiliation(s)
- Yu-Zheng He
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, No.215 Heping West Road, Shijiazhuang, Hebei, 050000, China
| | - Xiao-Ning Li
- Department of Thoracic Surgery, Hebei General Hospital, No. 348 Heping West Road, Shijiazhuang, Hebei, 050000, China
| | - Hai-Tao Li
- The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Respiratory Critical Care, Hebei Institute of Respiratory Diseases, No. 215 Heping West Road, Shijiazhuang, Hebei, 050000, China
| | - Xian-Hua Bai
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, Hebei, 050000, China
| | - Yan-Chao Liu
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, Hebei, 050000, China
| | - Fan-Nian Li
- Department of Thoracic Surgery, The First Hospital of XingTai, No.376 Shunde Road, XingTai City, Hebei Province, 054001, China
| | - Bao-Lei Lv
- Department of Thoracic Surgery, Shijiazhuang People's Hospital, No.365 Jianhua South Street, Shijiazhuang, 050000, Hebei Province, China
| | - Tian-Jie Qi
- The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Respiratory Critical Care, Hebei Institute of Respiratory Diseases, No. 215 Heping West Road, Shijiazhuang, Hebei, 050000, China
| | - Xiu-Min Zhao
- Department of The integrated treatment of traditional Chinese and Western Medicine, The Second Hospital of Hebei Medical University, No.215 Heping West Road, Shijiazhuang, Hebei, 050000, China
| | - Shuai Li
- The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Respiratory Critical Care, Hebei Institute of Respiratory Diseases, No. 215 Heping West Road, Shijiazhuang, Hebei, 050000, China.
| |
Collapse
|
24
|
Meng S, Wang C. Analysis of METTL14 expression in pancreatic cancer and adjacent tissues and its prognostic value for patient outcomes. Clin Exp Med 2024; 25:3. [PMID: 39527301 PMCID: PMC11554755 DOI: 10.1007/s10238-024-01506-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024]
Abstract
This study aims to analyze the differential expression of METTL14 in pancreatic cancer (PC) tissues and adjacent normal tissues, and its correlation with clinical outcomes. According to the inclusion and exclusion criteria, a total of 80 patients diagnosed in our hospital from January 2021 to January 2023 were chosen as research subjects. RTQ-PCR has detected the mRNA level expression of METTL14 in cancer and para-cancerous tissues. Immunohistochemistry was used to detect the protein expression of METTL14 in cancer and para-cancerous tissues. To compare the relationship between METTL14 expression and clinicopathological parameters in different PC patients. Kaplan-Meier survival analysis of the relationship between METTL14 expression in PC tissues and patient survival prognosis. The Multifactor COX model evaluates factors affecting the prognosis of PC. The expression level of METTL14 mRNA in PC tissues was 5.51 ± 0.35 (kDa), and the positive rate of METTL14 protein expression in PC tissues of all patients was 73.75 (59/80). Tumor location (P = 0.012), tumor differentiation degree (P = 0.028), tumor AJCC stage (P = 0.000), and lymph node metastasis (P = 0.000) were significantly related to the positive rate of METTL14 protein expression in PC tissue. Follow-up results showed that among 80 patients, 63 died. The three-year survival rate of the METTL14 positive group was 13.56% (8/59), and the three-year survival rate of the negative group was 42.86% (9/21). The difference in the three-year survival rate between METTL14 positive and negative expression groups was statistically significant (P = 0.031). Multivariate COX regression analysis results showed that METTL14 was positive (OR 2.797, 95% CI 1.233-5.877), tumor AJCC stage II-III (OR 1.628, 95% CI 1.435-3.859) and lymph node metastasis (OR 1.733, 95% CI 1.122-2.372) were substantive risk factors for poor prognosis in patients with PC. METTL14 expression increases in PC tissue, which is related to tumor AJCC stage, tumor differentiation, and lymph node metastasis, and can be evaluated in the survival prognosis of patients with PC.
Collapse
Affiliation(s)
- Siyu Meng
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110000, China
| | - Cong Wang
- Department of General Surgery, Shengjing Hospital Affiliated to China Medical University Shenbei Campus, No. 16 Puhe Avenue, Shenbei New District, Shenyang, 110000, Liaoning Province, China.
| |
Collapse
|
25
|
Poosri S, Boonyuen U, Chupeerach C, Soonthornworasiri N, Kwanbunjan K, Prangthip P. Association of FTO variants rs9939609 and rs1421085 with elevated sugar and fat consumption in adult obesity. Sci Rep 2024; 14:25618. [PMID: 39463443 PMCID: PMC11514288 DOI: 10.1038/s41598-024-77004-6] [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: 06/10/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024] Open
Abstract
This cross-sectional study explores the impact of FTO gene single nucleotide polymorphisms (SNPs) rs9939609 and rs1421085 on dietary habits contributing to obesity risk in Thai adults. The study enrolled 384 participants from Bangkok, categorized as non-obese (BMI < 25 kg/m2) or obese (BMI ≥ 25 kg/m2) based on WHO Asia Pacific Guidelines. Genotyping for FTO variants was performed using DNA from blood samples. While both SNPs adhered to Hardy-Weinberg equilibrium, the association between risk alleles and anthropometric measurements was not statistically significant. However, risk allele carriers showed significantly higher intakes of sugar and saturated fat compared to homozygous dominant individuals. In the obese group, the odds ratio for high-sugar intake was 2.22 (95% CI 1.13-4.37, p = 0.021) for rs9939609 risk allele carriers. For high-saturated fat intake, the odds ratio was 1.86 (95% CI 1.02-3.40, p = 0.041). Similar associations were observed for rs1421085. Risk allele carriers also exhibited significantly higher leptin levels (p < 0.043) and a positive correlation with myeloperoxidase levels (p < 0.038). These findings highlight the complex relationship between FTO risk alleles, increased consumption of sugar and saturated fat, and obesity-related parameters. The insights emphasize the importance of considering both genetic and dietary factors in obesity prevention strategies.
Collapse
Affiliation(s)
- Sakawrut Poosri
- Department of Tropical Nutrition and Food Science, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Usa Boonyuen
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Chaowanee Chupeerach
- Food and Nutrition Academic and Research Cluster, Institute of Nutrition, Mahidol University, Nakhon Pathom, Thailand
| | | | - Karunee Kwanbunjan
- Department of Tropical Nutrition and Food Science, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Pattaneeya Prangthip
- Department of Tropical Nutrition and Food Science, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
26
|
Hussain M, Waheed A, Elahi A, Mustafa G. Fat Mass and Obesity-Related (FTO) Gene Variant Is a Predictor of CVD in T2DM Patients. J Diabetes Res 2024; 2024:5914316. [PMID: 39257882 PMCID: PMC11383650 DOI: 10.1155/2024/5914316] [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: 01/16/2024] [Revised: 04/10/2024] [Accepted: 05/29/2024] [Indexed: 09/12/2024] Open
Abstract
Background: The role of the common FTO gene variant rs9939609 in obesity has been well established, and the FTO gene has a strong association with T2DM. Objective: To investigate the association of FTO gene variant rs9939609 with obesity-related parameters in T2DM and CVD patients. Materials and Methods: In this cross-sectional study, 280 subjects of either sex aged 45.10 ± 9.6 years were randomly divided into four groups, that is, T2DM, T2DM with CVD, nondiabetic with CVD disease, and normal control. These samples were genotyped by ARMS-PCR. The FTO gene association with obesity-related parameters in T2DM and CVD patients was analyzed by SPSS 22. Results: The TT genotype was the most common genotype (46.80%) in our study groups. The minor allele frequency (MAF) was significantly higher in T2DM patients (0.39 vs. 0.28), T2DM patients with CVD (0.43 vs. 0.28), and nondiabetic patients with CVD (0.35 vs. 0.28) as compared to control with p < 0.005. Carriers of the AA genotype of the FTO gene rs9939609 were significantly associated with increased BMI, WC, HbA1C, SBP, DBP, and TGs and lowered HDL cholesterol as compared to the TA and TT genotypes in T2DM and CVD patients with p < 0.005. The FTO gene variant rs9939609 showed a significant association with T2DM and CVD. The AA genotype odds ratio (OR) in T2DM was 1.48 (1.06-2.32), p = 0.006, and in CVD, it was 1.56 (1.04-2.4), p = 0.003. Conclusion: The FTO gene variant rs9939609 has a strong association with T2DM and CVD. The AA genotype of FTO gene variants rs9939609 showed a strong association with most of the risk factors of CVD and T2DM.
Collapse
Affiliation(s)
- Mazhar Hussain
- Pharmacology DepartmentSheikh Zayed Medical College, Rahim Yar Khan, Punjab, Pakistan
| | - Akbar Waheed
- Pharmacology DepartmentRiphah International University, Islamabad, Pakistan
| | - Asim Elahi
- Internal Medicine Residency ProgramSouth Texas Health GME ConsortiumTexas A & M School of Medicine, Bryan, Texas, USA
| | - Ghulam Mustafa
- Community Medicine DepartmentSheikh Zayed Medical College, Rahim Yar Khan, Punjab, Pakistan
| |
Collapse
|
27
|
Vranceanu M, Filip L, Hegheș SC, de Lorenzo D, Cozma-Petruț A, Ghitea TC, Stroia CM, Banc R, Mîrza OM, Miere D, Cozma V, Popa DS. Genes Involved in Susceptibility to Obesity and Emotional Eating Behavior in a Romanian Population. Nutrients 2024; 16:2652. [PMID: 39203789 PMCID: PMC11357152 DOI: 10.3390/nu16162652] [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: 07/15/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 09/03/2024] Open
Abstract
Obesity, a significant public health concern with high prevalence in both adults and children, is a complex disorder arising from the interaction of multiple genes and environmental factors. Advances in genome-wide association studies (GWAS) and sequencing technologies have identified numerous polygenic causes of obesity, particularly genes involved in hunger, satiety signals, adipocyte differentiation, and energy expenditure. This study investigates the relationship between six obesity-related genes (CLOCK, FTO, GHRL, LEP, LEPR, MC4R) and their impact on BMI, WC, HC, WHR, and emotional eating behavior in 220 Romanian adults. Emotional eating was assessed using the validated Emotional Eating Questionnaire (EEQ). Our analysis revealed significant variability in obesity-related phenotypes and emotional eating behaviors across different genotypes. Specifically, CLOCK/CC, FTO/AA, and LEP/AA genotypes were strongly associated with higher obesity metrics and emotional eating scores, while GHRL/TT and MC4R/CC were linked to increased BMI and WHR. The interplay between genetic predisposition and emotional eating behavior significantly influenced BMI and WHR, indicating a complex relationship between genetic and behavioral factors. This study, the first of its kind in Romania, provides a foundation for targeted interventions to prevent and reduce obesity and suggests potential strategies for gene expression modulation to mitigate the effects of emotional eating. Adopting a 'One Health' approach by creating an evidence base derived from both human and animal studies is crucial for understanding how to control obesity.
Collapse
Affiliation(s)
- Maria Vranceanu
- Department of Toxicology, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania (D.-S.P.)
| | - Lorena Filip
- Department of Bromatology, Hygiene, Nutrition, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania; (A.C.-P.); (R.B.); (O.M.M.); (D.M.)
- Academy of Romanian Scientists (AOSR), 3 Ilfov St, 050044 Bucharest, Romania
| | - Simona-Codruța Hegheș
- Department of Drug Analysis, Facullty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania;
| | - David de Lorenzo
- UCL Great Ormond Street Institute of Child Health, 30 Guilford St, London WC1N 1EH, UK;
| | - Anamaria Cozma-Petruț
- Department of Bromatology, Hygiene, Nutrition, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania; (A.C.-P.); (R.B.); (O.M.M.); (D.M.)
| | - Timea Claudia Ghitea
- Doctoral Scool of Biomedical Sciences, Faculty of Medicine and Pharmacy, University of Oradea, 1 Universităţii Street, 410087 Oradea, Romania; (T.C.G.)
| | - Carmina Mariana Stroia
- Doctoral Scool of Biomedical Sciences, Faculty of Medicine and Pharmacy, University of Oradea, 1 Universităţii Street, 410087 Oradea, Romania; (T.C.G.)
| | - Roxana Banc
- Department of Bromatology, Hygiene, Nutrition, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania; (A.C.-P.); (R.B.); (O.M.M.); (D.M.)
| | - Oana Maria Mîrza
- Department of Bromatology, Hygiene, Nutrition, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania; (A.C.-P.); (R.B.); (O.M.M.); (D.M.)
| | - Doina Miere
- Department of Bromatology, Hygiene, Nutrition, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania; (A.C.-P.); (R.B.); (O.M.M.); (D.M.)
| | - Vasile Cozma
- Department of Parasitology and Parasitic Diseases, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine of Cluj-Napoca, 3–5, Mănăştur Street, 400372 Cluj-Napoca, Romania;
- Academy of Agricultural and Forestry Sciences Gheorghe Ionescu-Siseşti (A.S.A.S.), 61, Mărăști Boulevard, 011464 Bucharest, Romania
| | - Daniela-Saveta Popa
- Department of Toxicology, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania (D.-S.P.)
| |
Collapse
|
28
|
Cucoreanu C, Tigu AB, Nistor M, Moldovan RC, Pralea IE, Iacobescu M, Iuga CA, Szabo R, Dindelegan GC, Ciuce C. Epigenetic and Molecular Alterations in Obesity: Linking CRP and DNA Methylation to Systemic Inflammation. Curr Issues Mol Biol 2024; 46:7430-7446. [PMID: 39057082 PMCID: PMC11275580 DOI: 10.3390/cimb46070441] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Obesity is marked by excessive fat accumulation in the adipose tissue, which disrupts metabolic processes and causes chronic systemic inflammation. Commonly, body mass index (BMI) is used to assess obesity-related risks, predicting potential metabolic disorders. However, for a better clustering of obese patients, we must consider molecular and epigenetic changes which may be responsible for inflammation and metabolic changes. Our study involved two groups of patients, obese and healthy donors, on which routine analysis were performed, focused on BMI, leukocytes count, and C-reactive protein (CRP) and completed with global DNA methylation and gene expression analysis for genes involved in inflammation and adipogenesis. Our results indicate that obese patients exhibited elevated leukocytes levels, along with increased BMI and CRP. The obese group revealed a global hypomethylation and upregulation of proinflammatory genes, with adipogenesis genes following the same trend of being overexpressed. The study confirms that obesity is linked to systematic inflammation and metabolic dysfunction through epigenetic and molecular alterations. The CRP was correlated with the hypomethylation status in obese patients, and this fact may contribute to a better understanding of the roles of specific genes in adipogenesis and inflammation, leading to a better personalized therapy.
Collapse
Affiliation(s)
- Ciprian Cucoreanu
- Department of General Surgery, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Adrian-Bogdan Tigu
- Department of Translational Medicine, Research Center for Advance Medicine—MEDFUTURE, “Iuliu Hațieganu” University of Medicine and Pharmacy Cluj-Napoca, 400012 Cluj-Napoca, Romania
| | - Madalina Nistor
- Department of Translational Medicine, Research Center for Advance Medicine—MEDFUTURE, “Iuliu Hațieganu” University of Medicine and Pharmacy Cluj-Napoca, 400012 Cluj-Napoca, Romania
| | - Radu-Cristian Moldovan
- Department of Proteomics and Metabolomics, Research Center for Advance Medicine—MEDFUTURE, “Iuliu Hațieganu” University of Medicine and Pharmacy Cluj-Napoca, 400012 Cluj-Napoca, Romania
| | - Ioana-Ecaterina Pralea
- Department of Proteomics and Metabolomics, Research Center for Advance Medicine—MEDFUTURE, “Iuliu Hațieganu” University of Medicine and Pharmacy Cluj-Napoca, 400012 Cluj-Napoca, Romania
- Department of Pharmaceutical Analysis, Faculty of Pharmacy, “Iuliu Haţieganu” University of Medicine and Pharmacy Cluj-Napoca, 400012 Cluj-Napoca, Romania
| | - Maria Iacobescu
- Department of Proteomics and Metabolomics, Research Center for Advance Medicine—MEDFUTURE, “Iuliu Hațieganu” University of Medicine and Pharmacy Cluj-Napoca, 400012 Cluj-Napoca, Romania
| | - Cristina-Adela Iuga
- Department of Proteomics and Metabolomics, Research Center for Advance Medicine—MEDFUTURE, “Iuliu Hațieganu” University of Medicine and Pharmacy Cluj-Napoca, 400012 Cluj-Napoca, Romania
- Department of Pharmaceutical Analysis, Faculty of Pharmacy, “Iuliu Haţieganu” University of Medicine and Pharmacy Cluj-Napoca, 400012 Cluj-Napoca, Romania
| | - Robert Szabo
- Department of Anesthesia and Intensive Care, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400012 Cluj-Napoca, Romania
| | - George-Calin Dindelegan
- Department of General Surgery, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Constatin Ciuce
- Department of General Surgery, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| |
Collapse
|
29
|
Steinwand S, Stacher Hörndli C, Ferris E, Emery J, Gonzalez Murcia JD, Cristina Rodriguez A, Leydsman TC, Chaix A, Thomas A, Davey C, Gregg C. Conserved Noncoding Cis-Elements Associated with Hibernation Modulate Metabolic and Behavioral Adaptations in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.26.600851. [PMID: 38979203 PMCID: PMC11230392 DOI: 10.1101/2024.06.26.600851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Our study elucidates functional roles for conserved cis-elements associated with the evolution of mammalian hibernation. Genomic analyses found topologically associated domains (TADs) that disproportionately accumulated convergent genomic changes in hibernators, including the TAD for the Fat Mass & Obesity (Fto) locus. Some hibernation-linked cis-elements in this TAD form regulatory contacts with multiple neighboring genes. Knockout mice for these cis-elements exhibit Fto, Irx3, and Irx5 gene expression changes, impacting hundreds of genes downstream. Profiles of pre-torpor, torpor, and post-torpor phenotypes found distinct roles for each cis-element in metabolic control, while a high caloric diet uncovered different obesogenic effects. One cis-element promoting a lean phenotype influences foraging behaviors throughout life, affecting specific behavioral sequences. Thus, convergent evolution in hibernators pinpoints functional genetic mechanisms of mammalian metabolic control.
Collapse
Affiliation(s)
- Susan Steinwand
- Department of Neurobiology, University of Utah; Salt Lake City, 84105, USA
| | | | - Elliott Ferris
- Department of Neurobiology, University of Utah; Salt Lake City, 84105, USA
| | - Jared Emery
- Department of Neurobiology, University of Utah; Salt Lake City, 84105, USA
| | | | | | - Tyler C. Leydsman
- Department of Neurobiology, University of Utah; Salt Lake City, 84105, USA
| | - Amandine Chaix
- Department of Nutrition and Integrative Physiology, University of Utah; Salt Lake City, 84105, USA
| | - Alun Thomas
- Division of Epidemiology, University of Utah; Salt Lake City, 84105, USA
- Study Design and Biostatistics Center, University of Utah; Salt Lake City, 84105, USA
| | - Crystal Davey
- Mutation Generation & Detection Core Facility, University of Utah; Salt Lake City, 84105, USA
| | - Christopher Gregg
- Department of Neurobiology, University of Utah; Salt Lake City, 84105, USA
- Department of Human Genetics, University of Utah; Salt Lake City, 84105, USA
| |
Collapse
|
30
|
Chandra Sekar PK, Veerabathiran R. Genes linked to obesity-related infertility: bridging the knowledge gap. REPRODUCTIVE AND DEVELOPMENTAL MEDICINE 2024; 8:121-129. [DOI: 10.1097/rd9.0000000000000096] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025] Open
Abstract
Genetic factors play a pivotal role in the complex relationship between obesity and infertility. This article delves into the genetics of obesity-related infertility, focusing on the essential genes and mechanisms in both sexes. We explored infertility factors in obese females, focusing on polycystic ovary syndrome (PCOS) and the influence of genes like insulin receptor (INSR), androgen receptor (AR), and follicle-stimulating hormone receptor (FSHR). Epigenetic changes are believed to contribute to PCOS-related infertility. The impact of adipokines and inflammation on obesity-related infertility has been discussed, with genes such as fat mass and obesity (FTO) and melanocortin-4-receptor (MC4R) playing significant roles. Genetic factors affecting sperm quality and function, including nuclear receptor subfamily 3 group C member 1 (NR3C1) and methylenetetrahydrofolate reductase (MTHFR), have been investigated in obesity-related infertility in males. Hormonal dysregulation influenced by genetic markers, such as leptin receptor (LEPR), tumor necrosis factor-alpha (TNF-α), and interleukin-6 (IL-6), was also examined. Genetic factors play a vital role in obesity-related infertility in both sexes. Genes involved in metabolism, hormonal regulation, and inflammation contribute to the complex association between obesity and infertility. Epigenetic changes further complicate the relationship. Understanding these genetic mechanisms is essential to address obesity-related infertility and develop personalized interventions.
Collapse
Affiliation(s)
- Praveen Kumar Chandra Sekar
- Human Cytogenetics and Genomics Laboratory, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu 603103, India
| | - Ramakrishnan Veerabathiran
- Human Cytogenetics and Genomics Laboratory, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu 603103, India
| |
Collapse
|
31
|
Khademi Z, Mahmoudi Z, Sukhorukov VN, Jamialahmadi T, Sahebkar A. CRISPR/Cas9 Technology: A Novel Approach to Obesity Research. Curr Pharm Des 2024; 30:1791-1803. [PMID: 38818919 DOI: 10.2174/0113816128301465240517065848] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 04/11/2024] [Accepted: 04/17/2024] [Indexed: 06/01/2024]
Abstract
Gene editing technology, particularly Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) has transformed medical research. As a newly developed genome editing technique, CRISPR technology has strongly assisted scientists in enriching their comprehension of the roles of individual genes and their influences on a vast spectrum of human malignancies. Despite considerable progress in elucidating obesity's molecular pathways, current anti-obesity medications fall short in effectiveness. A thorough understanding of the genetic foundations underlying various neurobiological pathways related to obesity, as well as the neuro-molecular mechanisms involved, is crucial for developing effective obesity treatments. Utilizing CRISPR-based technologies enables precise determination of the roles of genes that encode transcription factors or enzymes involved in processes, such as lipogenesis, lipolysis, glucose metabolism, and lipid storage within adipose tissue. This innovative approach allows for the targeted suppression or activation of genes regulating obesity, potentially leading to effective weight management strategies. In this review, we have provided a detailed overview of obesity's molecular genetics, the fundamentals of CRISPR/Cas9 technology, and how this technology contributes to the discovery and therapeutic targeting of new genes associated with obesity.
Collapse
Affiliation(s)
- Zahra Khademi
- Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Zahra Mahmoudi
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Vasily N Sukhorukov
- Institute of General Pathology and Pathophysiology, The Russian Academy of Medical Sciences, 8 Baltiiskaya Street, Moscow 125315, Russia
| | - Tannaz Jamialahmadi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
32
|
Conery M, Grant SFA. Human height: a model common complex trait. Ann Hum Biol 2023; 50:258-266. [PMID: 37343163 PMCID: PMC10368389 DOI: 10.1080/03014460.2023.2215546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 04/10/2023] [Accepted: 05/09/2023] [Indexed: 06/23/2023]
Abstract
CONTEXT Like other complex phenotypes, human height reflects a combination of environmental and genetic factors, but is notable for being exceptionally easy to measure. Height has therefore been commonly used to make observations later generalised to other phenotypes though the appropriateness of such generalisations is not always considered. OBJECTIVES We aimed to assess height's suitability as a model for other complex phenotypes and review recent advances in height genetics with regard to their implications for complex phenotypes more broadly. METHODS We conducted a comprehensive literature search in PubMed and Google Scholar for articles relevant to the genetics of height and its comparatibility to other phenotypes. RESULTS Height is broadly similar to other phenotypes apart from its high heritability and ease of measurment. Recent genome-wide association studies (GWAS) have identified over 12,000 independent signals associated with height and saturated height's common single nucleotide polymorphism based heritability of height within a subset of the genome in individuals similar to European reference populations. CONCLUSIONS Given the similarity of height to other complex traits, the saturation of GWAS's ability to discover additional height-associated variants signals potential limitations to the omnigenic model of complex-phenotype inheritance, indicating the likely future power of polygenic scores and risk scores, and highlights the increasing need for large-scale variant-to-gene mapping efforts.
Collapse
Affiliation(s)
- Mitchell Conery
- Division of Human Genetics, Center for Spatial and Functional Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine at the University of PA, Philadelphia, PA, USA
- Department of Pharmacology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Struan F A Grant
- Division of Human Genetics, Center for Spatial and Functional Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine at the University of PA, Philadelphia, PA, USA
- Division of Diabetes and Endocrinology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
33
|
Hoseini Tavassol Z, Mousavi SM, Molaei B, Bandarian F, Ejtahed HS, Khalagi K, Ghannadi S, Larijani B, Hasani-Ranjbar S. Association of fat mass and obesity-associated ( FTO) gene polymorphisms with non-communicable diseases (NCDs) in the Iranian population: A systematic review of observational studies. J Diabetes Metab Disord 2022; 21:1975-1989. [PMID: 36404828 PMCID: PMC9672241 DOI: 10.1007/s40200-022-01139-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 10/03/2022] [Indexed: 10/31/2022]
Abstract
Background Single nucleotide polymorphisms have been implicated in various diseases, most notably non-communicable diseases (NCDs). The aim of this study was to review available evidence regarding associations between FTO polymorphisms and NCDs in the Iranian population. Methods A comprehensive search was conducted through PubMed/Medline and Scopus databases up to December 2021, as well as reference lists of pertinent articles and key journals. All observational studies that examined the association between FTO gene polymorphisms and NCDs in the Iranian population were included. There was no limitation on the publication year. The Newcastle-Ottawa Scale (NOS) was used to assess the study's quality. Results The initial search yielded 95 studies, of which 30 studies were included in the current systematic review. The underlying disorders were obesity, type 2 diabetes, breast and colorectal cancers, depression, and metabolic syndrome. These studies found an association between FTO gene polymorphisms and obesity in the Iranian population, but the relationship with other NCDs was debatable. Even though, other diseases such as diabetes and metabolic syndrome, which are closely related to obesity, may also be associated with FTO gene polymorphisms. Conclusion FTO gene polymorphism appears to play a role in the occurrence of NCDs. Some of the study results may be misleading due to ethnic differences and the effect of other genetic factors on disease onset, which needs to be investigated further. Finally, FTO gene polymorphisms can be studied as a preventive or therapeutic target. Supplementary Information The online version contains supplementary material available at 10.1007/s40200-022-01139-4.
Collapse
Affiliation(s)
- Zahra Hoseini Tavassol
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Mousavi
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahareh Molaei
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Bandarian
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institutes, Tehran University of Medical Sciences, Tehran, Iran
| | - Hanieh-Sadat Ejtahed
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Kazem Khalagi
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Osteoporosis Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shima Ghannadi
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Sports Medicine Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shirin Hasani-Ranjbar
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|