1
|
Liu J, Ma C, Cheng Y, Wang M, Zhao G, Huang L, Song R, Wang X, Li H. METTL14 and WTAP play a crucial role in the regulation of bovine preadipocyte differentiation. Anim Biotechnol 2025; 36:2476531. [PMID: 40094566 DOI: 10.1080/10495398.2025.2476531] [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: 10/23/2024] [Accepted: 03/03/2025] [Indexed: 03/19/2025]
Abstract
m6A methylation is the most common mRNA modification in mammals and plays a significant role in regulating various biological functions. Some studies have demonstrated that the methyltransferase METTL3 can promote adipogenesis. However, the regulatory mechanisms of METTL14 and WTAP, both methyltransferases, in adipogenesis remain unclear. This study investigated their effects on bovine preadipocyte differentiation using siRNA-mediated knockdown combined with transcriptomic analysis. Silencing METTL14 and WTAP significantly impaired lipid droplet formation and revealed distinct regulatory pathways: METTL14 knockdown affected genes like JAK2 and STAT3, while WTAP suppression down-regulated PPARγ/FABP4 signalling pathway components. These findings demonstrate that WTAP specifically modulates bovine adipocyte differentiation through the PPARγ/FABP4 pathway.
Collapse
Affiliation(s)
- Jia Liu
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Chicheng Ma
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Yu Cheng
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Minzhi Wang
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Guoqing Zhao
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Liwei Huang
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Ruigao Song
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
- Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - Xi Wang
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Hongxia Li
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
2
|
Yang B, Wang M, Wu Z, Tan J, Meng Y, Zhang T, Zan L, Yang W. Perilipin1 mediates milk fat synthesis in bovine mammary epithelial cells through SREBP1 phosphorylation. Anim Biotechnol 2025; 36:2497915. [PMID: 40338730 DOI: 10.1080/10495398.2025.2497915] [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: 02/27/2025] [Accepted: 04/21/2025] [Indexed: 05/10/2025]
Abstract
This study investigates the role of Perilipin1 (PLIN1) in milk fat synthesis in bovine mammary epithelial cells (BMECs) and its regulatory mechanisms, aiming to provide a foundation for improving milk fat content through molecular breeding. BMECs were used as a model to analyze the effects of PLIN1 overexpression (OE-PLIN1) and interference (si-PLIN1) on milk fat synthesis and lipid-related gene expression using RT-qPCR, Western blot, and Oil Red O staining. Results show that OE-PLIN1 significantly enhances triglyceride (TAG) accumulation in BMECs (P < 0.01), upregulates lipid synthesis-related genes (such as PPARγ, C/EBPα, C/EBPβ, FABP4, FASN) (P < 0.05), and downregulates the mRNA expression of lipid breakdown-related genes (HSL, ATGL) (P < 0.05). Conversely, si-PLIN1 significantly reduces TAG accumulation (P < 0.05) and lowers the expression of lipid synthesis and breakdown genes (P < 0.05). Additionally, OE-PLIN1 combined with SREBP1 siRNA interference (si-SREBP1) did not have a significant impact on the mRNA and protein levels of SREBP1, but it significantly altered SREBP1's phosphorylation, indicating that SREBP1 interference inhibits PLIN1's effect on milk fat synthesis. This study suggests that PLIN1 promotes milk fat synthesis in BMECs via regulating SREBP1 activity, offering a new strategy for enhancing milk fat content in dairy cattle.
Collapse
Affiliation(s)
- Benshun Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Meng Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Zhangqing Wu
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Jianbing Tan
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yanhong Meng
- Shaanxi Qinbao Animal Husbandry Co., LTD, Yangling, China
| | - Taoping Zhang
- Shaanxi Qinbao Animal Husbandry Co., LTD, Yangling, China
| | - Linsen Zan
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Wucai Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
- Northwest A&F University Shenzhen Research Institute, Shenzhen, China
| |
Collapse
|
3
|
Nicchio IG, Cirelli T, Quil LCDC, Camilli AC, Scarel-Caminaga RM, Leite FRM. Understanding the peroxisome proliferator-activated receptor gamma (PPAR-γ) role in periodontitis and diabetes mellitus: A molecular perspective. Biochem Pharmacol 2025; 237:116908. [PMID: 40157459 DOI: 10.1016/j.bcp.2025.116908] [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: 12/12/2024] [Revised: 02/19/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Periodontitis and Type 2 Diabetes Mellitus (T2DM) are chronic conditions with dysregulated immune responses. Periodontitis involves immune dysfunction and dysbiotic biofilms, leading to tissue destruction. T2DM is marked by insulin resistance and systemic inflammation, driving metabolic and tissue damage. Both conditions share activation of key pathways, including Nuclear Factor Kappa B (NF-κB), Activator Protein-1 (AP-1), and Signal Transducer and Activator of Transcription (STAT) proteins, reinforcing an inflammatory feedback loop. This review highlights the role of Peroxisome Proliferator-Activated Receptor Gamma (PPAR-γ), a transcription factor central to lipid and glucose metabolism, adipogenesis, and immune regulation. PPAR-γ activation has been shown to suppress inflammatory mediators such as Tumor Necrosis Factor Alpha (TNF-α) and Interleukin 6 (IL-6) through the inhibition of NF-κB, AP-1, and STAT pathways, thereby potentially disrupting the inflammatory-metabolic cycle that drives both diseases. PPAR-γ agonists, including thiazolidinediones (TZDs) and endogenous ligands such as 15-deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2), show promise in reducing inflammation and improving insulin sensitivity, but they are limited by adverse effects. Therapies, including Selective Peroxisome Proliferator-Activated Receptor Modulators (SPPARMs), have been developed to offer a more targeted approach, allowing for selective modulation of PPAR-γ activity to retain its anti-inflammatory benefits while minimizing their side effects. By integrating insights into PPAR-γ's molecular mechanisms, this review underscores its therapeutic potential in mitigating inflammation and enhancing metabolic control.
Collapse
Affiliation(s)
- Ingra Gagno Nicchio
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara, São Paulo State University-UNESP, Araraquara 14801-903, SP, Brazil; Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, School of Dentistry at Araraquara, São Paulo State University-UNESP, Araraquara 14801-903, SP, Brazil.
| | - Thamiris Cirelli
- Department of Dentistry, Centro Universitário das Faculdades Associadas, São João da Boa Vista 13870-377, SP, Brazil.
| | - Lucas César da Costa Quil
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara, São Paulo State University-UNESP, Araraquara 14801-903, SP, Brazil; Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, School of Dentistry at Araraquara, São Paulo State University-UNESP, Araraquara 14801-903, SP, Brazil.
| | - Angelo Constantino Camilli
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara, São Paulo State University-UNESP, Araraquara 14801-903, SP, Brazil.
| | - Raquel Mantuaneli Scarel-Caminaga
- Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, School of Dentistry at Araraquara, São Paulo State University-UNESP, Araraquara 14801-903, SP, Brazil.
| | - Fabio Renato Manzolli Leite
- National Dental Research Institute Singapore, National Dental Centre Singapore, 168938, Singapore; Oral Health Academic Clinical Programme, Duke-NUS Medical School, 169857, Singapore.
| |
Collapse
|
4
|
Kiani P, Khodadadi ES, Nikdasti A, Yarahmadi S, Gheibi M, Yousefi Z, Ehtiati S, Yahyazadeh S, Shafiee SM, Taghizadeh M, Igder S, Khatami SH, Karima S, Vakili O, Pourfarzam M. Autophagy and the peroxisome proliferator-activated receptor signaling pathway: A molecular ballet in lipid metabolism and homeostasis. Mol Cell Biochem 2025; 480:3477-3499. [PMID: 39891864 DOI: 10.1007/s11010-025-05207-0] [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: 10/11/2024] [Accepted: 01/04/2025] [Indexed: 02/03/2025]
Abstract
Lipids, which are indispensable for cellular architecture and energy storage, predominantly consist of triglycerides (TGs), phospholipids, cholesterol, and their derivatives. These hydrophobic entities are housed within dynamic lipid droplets (LDs), which expand and contract in response to nutrient availability. Historically perceived as a cellular waste disposal mechanism, autophagy has now been recognized as a crucial regulator of metabolism. Within this framework, lipophagy, the selective degradation of LDs, plays a fundamental role in maintaining lipid homeostasis. Dysregulated lipid metabolism and autophagy are frequently associated with metabolic disorders such as obesity and atherosclerosis. In this context, peroxisome proliferator-activated receptors (PPARs), particularly PPAR-γ, serve as intracellular lipid sensors and master regulators of gene expression. Their regulatory influence extends to both autophagy and lipid metabolism, indicating a complex interplay between these processes. This review explores the hypothesis that PPARs may directly modulate autophagy within the realm of lipid metabolism, thereby contributing to the pathogenesis of metabolic diseases. By elucidating the underlying molecular mechanisms, we aim to provide a comprehensive understanding of the intricate regulatory network that connects PPARs, autophagy, and lipid homeostasis. The crosstalk between PPARs and other signaling pathways underscores the complexity of their regulatory functions and the potential for therapeutic interventions targeting these pathways. The intricate relationships among PPARs, autophagy, and lipid metabolism represent a pivotal area of research with significant implications for understanding and treating metabolic disorders.
Collapse
Affiliation(s)
- Pouria Kiani
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elaheh Sadat Khodadadi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122, Padova, Italy
| | - Ali Nikdasti
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020, Legnaro, Padova, Italy
| | - Sahar Yarahmadi
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mobina Gheibi
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zeynab Yousefi
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sajad Ehtiati
- Student Research Committee, Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sheida Yahyazadeh
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sayed Mohammad Shafiee
- Autophagy Research Center, Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Motahareh Taghizadeh
- Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Somayeh Igder
- Department of Clinical Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyyed Hossein Khatami
- Student Research Committee, Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Saeed Karima
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran.
| | - Omid Vakili
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Morteza Pourfarzam
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
5
|
Kaimala S, Yassin LK, Hamad MIK, Allouh MZ, Sampath P, AlKaabi J, Khee-Shing Leow M, Shehab S, Ansari SA, Emerald BS. Epigenetic crossroads in metabolic and cardiovascular health: the role of DNA methylation in type 2 diabetes and cardiovascular diseases. Cardiovasc Diabetol 2025; 24:231. [PMID: 40442704 PMCID: PMC12124063 DOI: 10.1186/s12933-025-02800-x] [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] [Received: 03/31/2025] [Accepted: 05/20/2025] [Indexed: 06/02/2025] Open
Abstract
Type 2 diabetes (T2D) and cardiovascular diseases (CVD), part of the metabolic syndrome (MetS), are major contributors to the global health crisis today. A recent report from the World Health Organisation estimates that 17.9 million lives are lost each year to CVD, and one-third of these are premature. The international diabetes federation estimates that around 537 million adults aged between 20 and 79 years are living with diabetes. People with diabetes are suggested to have twice the risk of developing CVD. Epigenetic modifications are being increasingly recognised as the key mediators linking genetic and environmental conditions to metabolic dysfunction. Among these, DNA methylation plays a crucial role in modulating gene expression and influencing pathways involved in glucose homeostasis, inflammation, and vascular integrity. Despite the advances in our understanding of the role of epigenetic alterations in metabolic diseases, including that of T2D, the mechanisms driving selective methylation changes and their long-term impact on cardiovascular health are still not well understood. This review synthesises the current knowledge on DNA methylation dynamics in T2D and their role towards the progression of CVD and explores their potential as biomarkers and therapeutic targets. Understanding the interplay between metabolism and epigenetics in the pathogenesis of T2D and CVD could provide critical insights for early disease identification and the development of novel epigenome-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Suneesh Kaimala
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, Abu Dhabi, United Arab Emirates
| | - Lidya K Yassin
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, Abu Dhabi, United Arab Emirates
| | - Mohammad I K Hamad
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, Abu Dhabi, United Arab Emirates
| | - Mohammed Z Allouh
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, Abu Dhabi, United Arab Emirates
| | - Prabha Sampath
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Juma AlKaabi
- Department of Internal Medicine, College of Medicine and Health Sciences, UAE University, P.O. Box 15551, Al Ain, United Arab Emirates
| | - Melvin Khee-Shing Leow
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), 30 Medical Drive, Singapore, 117609, Singapore
- Singapore Institute of Food and Biotechnology Innovation, A*STAR, Singapore, Singapore
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore, Singapore
- Cardiovascular and Metabolic Diseases Program, Duke-NUS Medical School, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Safa Shehab
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, Abu Dhabi, United Arab Emirates
| | - Suraiya Anjum Ansari
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, UAE
- Zayed Center for Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, Abu Dhabi, United Arab Emirates
- ASPIRE Precision Medicine, Research Institute Abu Dhabi, P.O. Box 15551, Al Ain, Abu Dhabi, United Arab Emirates
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, Abu Dhabi, United Arab Emirates.
- Zayed Center for Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, Abu Dhabi, United Arab Emirates.
- ASPIRE Precision Medicine, Research Institute Abu Dhabi, P.O. Box 15551, Al Ain, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
6
|
Cha SE, Lee MN, Kim ES. Metabolic impacts of long-chain fatty acids on cardiomyocyte maturation in neonatal mammalian hearts. Methods 2025; 241:S1046-2023(25)00131-8. [PMID: 40449856 DOI: 10.1016/j.ymeth.2025.05.010] [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: 03/09/2025] [Revised: 05/23/2025] [Accepted: 05/25/2025] [Indexed: 06/03/2025] Open
Abstract
Cardiomyocytes are essential models for cardiac disease modeling, drug development, and regenerative therapies. Specifically, human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have emerged as widely used cellular models with high reproducibility. However, cardiomyocytes generated in vitro tend to remain immature and insufficient in replicating the electrophysiological and mechanical functions of adult cardiomyocytes, limiting the clinical and experimental applications of these models. Thus, various biochemical and biophysical strategies have been explored to promote the maturation of cardiomyocytes, to address these limitations, and more accurately mimic the characteristics of mature cardiomyocytes. This review summarizes recent studies on multiple methodologies employed to induce cardiomyocyte maturation, with a particular emphasis on the role of long-chain fatty acids (LCFAs). The evidence summarized in this review is derived from studies utilizing cardiomyocytes from neonatal mice or rats and hiPSC-CMs. Meanwhile, immature cardiomyocytes have been demonstrated to predominantly rely on glycolysis, transitioning to oxidative phosphorylation through maturation, which enhances electrical stability, contractility, and structural organization. LCFAs play a key role in the cardiomyocyte maturation process by serving as key metabolic factors that generate ATP through mitochondrial β-oxidation, thereby improving metabolic efficiency. Additionally, LCFAs are involved in activating cytoskeletal components and signaling pathways integral to cardiomyocyte contractility. Importantly, studies suggest that when multiple biochemical and biophysical stimuli are simultaneously applied, various aspects of cardiomyocyte maturation are synergistically accelerated. Therefore, future studies focusing on the coordinated application of these regulatory factors are expected to enhance the maturation process, ultimately contributing to the generation of mature cardiomyocytes suitable for regenerative medicine and other advanced applications.
Collapse
Affiliation(s)
- Seong-Eung Cha
- Department of Biological Sciences and Biotechnology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Mi Nam Lee
- Department of Biological Sciences, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Eung-Sam Kim
- Department of Biological Sciences and Biotechnology, Chonnam National University, Gwangju 61186, Republic of Korea; Department of Biological Sciences, Chonnam National University, Gwangju 61186, Republic of Korea; Research Center of Ecomimetics, Institute of Sustainable Ecological Environment, Chonnam National University, Gwangju 61186, Republic of Korea; Center for Next-Generation Sensor Research and Development, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
7
|
Amin SA, Chakraborty G, Tarafdar R, Sessa L, Das I, Piotto S. Structural insights and molecular profiling of a large set of diverse compounds targeting PPARγ: from comprehensive cheminformatics approach to tool development. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2025; 36:443-461. [PMID: 40493266 DOI: 10.1080/1062936x.2025.2514061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Accepted: 05/26/2025] [Indexed: 06/12/2025]
Abstract
This study integrates a robust cheminformatics approach (including chemical space exploration, Bayesian model-based fingerprint analysis, and cluster-driven molecular profiling) to reveal the key structural features influencing peroxisome proliferator activated receptor-gamma (PPARγ) modulatory activity. The Bayesian classification model effectively differentiates between the beneficial and adverse structural characteristics of PPARγ modulators. Structural motifs such as substituted benzylamine, phenoxyphenyl groups, 5-phenyl-1,3-thiazolidine scaffolds, and indole rings have been identified as positive contributors, enhancing PPARγ activity. Conversely, features like substituted tertiary amines and sulphonamide groups were found to have detrimental effects, suggesting that these should be deprioritized in the design of future PPARγ modulators. Additionally, molecular clustering provided a means to categorize structurally similar compounds, facilitating scaffold analysis, diversity calculation, and lead optimization for PPARγ modulators. To extend these findings to the broader scientific community, we have developed an open-access online tool, 'Fasda_v1.0', (https://fasdav1web.streamlit.app/) designed for cluster-driven molecular profiling of any dataset, enabling further exploration and application of these methods. This study offers valuable guidance for designing and developing novel therapeutics targeting PPARγ, thereby contributing to advancements in treating type 2 diabetes mellitus and related diseases.
Collapse
Affiliation(s)
- S A Amin
- Department of Pharmacy, University of Salerno, Fisciano, Italy
- Department of Pharmaceutical Technology, JIS University, Kolkata, India
| | - G Chakraborty
- Department of Pharmaceutical Technology, JIS University, Kolkata, India
- Department of Pharmaceutical Chemistry, Eminent college of Pharmaceutical Technology, Barasat, India
| | - R Tarafdar
- Department of Pharmaceutical Technology, JIS University, Kolkata, India
| | - L Sessa
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| | - I Das
- Department of Pharmaceutical Technology, JIS University, Kolkata, India
| | - S Piotto
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| |
Collapse
|
8
|
Yuan P, Li H, Zhang H, Fan S, Dai Y, Jia J, Shen J, Zhang Y, Li H, Sun G, Liu X, Tian Y, Kang X, Zhao Y, Li G. miR-200b-3p affects the proliferation and differentiation of chicken preadipocytes by modulating SESN1 expression through competition with CircADGRF5. Poult Sci 2025; 104:105068. [PMID: 40132317 PMCID: PMC11984591 DOI: 10.1016/j.psj.2025.105068] [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: 12/02/2024] [Revised: 03/01/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025] Open
Abstract
Excessive deposition of abdominal fat in chickens has a significant impact on the poultry industry, and there is increasing evidence that non-coding RNAs play a crucial role in fat development. In our previous RNA-seq study, miR-200b-3p was found to be differentially expressed during different developmental periods of fat in Gushi chickens. In this study, we report that miR-200b-3p can directly bind to the 3'UTR region of SESN1 to promote proliferation and inhibit differentiation of preadipocytes. Overexpression of SESN1 inhibits preadipocyte proliferation and promotes differentiation. In contrast, inhibition of SESN1 resulted in the opposite outcome. Interestingly, we also identified the circADGRF5/miR-200b-3p/SESN1 ceRNA network involved in the developmental process of preadipocytes. Furthermore, we validated a novel circRNA, circADGRF5, in this report and found that it regulates SESN1 expression through competitive binding with miR-200b-3p. In conclusion, these data suggest that miR-200b-3p directly targets SESN1 to regulate the proliferation and differentiation of preadipocytes, and circADGRF5 regulates SESN1 expression through competitive binding with miR-200b-3p.
Collapse
Affiliation(s)
- Pengtao Yuan
- The Shennong Laboratory, Henan Agricultural University, Zheng Zhou, Henan Province, 450046, PR China
| | - Hongtai Li
- The Shennong Laboratory, Henan Agricultural University, Zheng Zhou, Henan Province, 450046, PR China
| | - Hongyuan Zhang
- The Shennong Laboratory, Henan Agricultural University, Zheng Zhou, Henan Province, 450046, PR China
| | - Shengxin Fan
- The Shennong Laboratory, Henan Agricultural University, Zheng Zhou, Henan Province, 450046, PR China
| | - Yaqi Dai
- The Shennong Laboratory, Henan Agricultural University, Zheng Zhou, Henan Province, 450046, PR China
| | - Jiyu Jia
- The Shennong Laboratory, Henan Agricultural University, Zheng Zhou, Henan Province, 450046, PR China
| | - Jingqi Shen
- The Shennong Laboratory, Henan Agricultural University, Zheng Zhou, Henan Province, 450046, PR China
| | - Yanhua Zhang
- The Shennong Laboratory, Henan Agricultural University, Zheng Zhou, Henan Province, 450046, PR China
| | - Hong Li
- The Shennong Laboratory, Henan Agricultural University, Zheng Zhou, Henan Province, 450046, PR China
| | - Guirong Sun
- The Shennong Laboratory, Henan Agricultural University, Zheng Zhou, Henan Province, 450046, PR China
| | - Xiaojun Liu
- The Shennong Laboratory, Henan Agricultural University, Zheng Zhou, Henan Province, 450046, PR China
| | - Yadong Tian
- The Shennong Laboratory, Henan Agricultural University, Zheng Zhou, Henan Province, 450046, PR China
| | - Xiangtao Kang
- The Shennong Laboratory, Henan Agricultural University, Zheng Zhou, Henan Province, 450046, PR China
| | - Yinli Zhao
- College of Biological Engineering, Henan University of Technology, Zheng Zhou, Henan Province, 450001, PR China.
| | - Guoxi Li
- The Shennong Laboratory, Henan Agricultural University, Zheng Zhou, Henan Province, 450046, PR China.
| |
Collapse
|
9
|
Liu J, Deng L, Yao B, Zhang Y, Huang J, Huang S, Liang C, Shen Y, Wang X. Carboxylesterase 2A gene knockout or enzyme inhibition alleviates steatohepatitis in rats by regulating PPARγ and endoplasmic reticulum stress. Free Radic Biol Med 2025; 232:279-291. [PMID: 40089078 DOI: 10.1016/j.freeradbiomed.2025.03.021] [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: 01/14/2025] [Revised: 03/09/2025] [Accepted: 03/13/2025] [Indexed: 03/17/2025]
Abstract
Metabolic dysfunction associated steatotic liver disease (MASLD) is a widespread liver disease that progresses from simple steatosis to severe steatohepatitis stage. Despite the recognized importance of carboxylesterase 2 (CES2) in hepatic lipid metabolism, the role of CES2 in hepatic inflammation remains unclear. The rat genome encodes six Ces2 genes and Ces2a shows high expression in the liver and intestine. Lipid metabolism, inflammation, fibrosis, and endoplasmic reticulum (ER) stress were investigated in Ces2a knockout (KO) rats. KO rats showed spontaneous liver lipid accumulation due to increased lipogenesis and reduced fatty acid oxidation. Non-targeted lipidomic analysis revealed enhanced lysophosphatidylcholines (LPCs) and phosphatidylcholines (PCs) in KO rats and increased concentrations of ligands, thus activating the expression of PPARγ. Although there was simple lipid accumulation in the liver of KO rats, Ces2a deficiency showed a significant protective effect against LPS and diet-induced hepatic steatohepatitis by inhibiting ER stress regulated by PPARγ activation. In line with this, treatment with tanshinone IIA, a CES2 inhibitor, significantly alleviated the progression of steatohepatitis induced by the MCD diet. In conclusion, the increased PPARγ expression in Ces2a deficiency may counteract liver inflammation and ER stress despite the presence of simple steatosis. Therefore, CES2 inhibition represents a potential therapeutic approach for steatohepatitis.
Collapse
Affiliation(s)
- Jie Liu
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Luyao Deng
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Bingyi Yao
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Yuanjin Zhang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Junze Huang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Shengbo Huang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Chenmeizi Liang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Yifei Shen
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Xin Wang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China.
| |
Collapse
|
10
|
Grytten E, Laupsa-Borge J, Cetin K, Bohov P, Nordrehaug JE, Skorve J, Berge RK, Strand E, Bjørndal B, Nygård OK, Rostrup E, Mellgren G, Dankel SN. Inflammatory markers after supplementation with marine n-3 or plant n-6 PUFAs: A randomized double-blind crossover study. J Lipid Res 2025; 66:100770. [PMID: 40058591 PMCID: PMC11999210 DOI: 10.1016/j.jlr.2025.100770] [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: 10/06/2022] [Revised: 03/03/2025] [Accepted: 03/06/2025] [Indexed: 04/04/2025] Open
Abstract
Omega-3 (n-3) (e.g., EPA/DHA) and omega-6 (n-6) (e.g., linoleic acid [LA]) FAs are suggested to have opposite effects on inflammation, but results are inconsistent and direct comparisons of n-3 and n-6 are lacking. In a double-blind, randomized, and crossover study, females (n = 16) and males (n = 23) aged 30-70 years with abdominal obesity were supplemented with 3-4 g/d EPA/DHA (fish oil) or 15-20 g/d LA (safflower oil) for 7 weeks, with a 9-week washout phase. Cytokines and chemokines (multiplex assay), acute-phase proteins (MALDI-TOF mass spectrometry), endothelial function (vascular reaction index), blood pressure, FA composition (red blood cell membranes/serum/adipose tissue, GC-MS/MS), and adipose gene expression (microarrays, quantitative PCR) were measured. While significant differences between treatments in relative change scores were found for systolic blood pressure (n-3 vs. n-6: -1.81% vs. 2.61%, P = 0.003), no differences between n-3 and n-6 were found for any circulatory inflammatory markers. However, compared with baseline, n-3 was followed by reductions in circulating TNF (-24.9%, P < 0.001), regulated upon activation, normal T cell expressed and secreted (-12.1%, P < 0.001), and macrophage inflammatory protein 1-beta (-12.5%, P = 0.014), and n-6 by lowered TNF (-18.8%, P < 0.001), regulated upon activation, normal T cell expressed and secreted (-7.37%, P = 0.027), monocyte chemoattractant protein-1 (-7.81%, P = 0.020), and macrophage inflammatory protein 1-beta (-14.2%, P = 0.010). Adipose tissue showed significant treatment differences in weight percent of EPA (n-3 vs. n-6: 50.2%∗ vs. -1.38%, P < 0.001, ∗: significant within-treatment change score), DHA (16.0%∗ vs. -3.67%, P < 0.001), and LA (-0.033 vs. 4.91%∗, P < 0.001). Adipose transcriptomics revealed overall downregulation of genes related to inflammatory processes after n-3 and upregulation after n-6, partly correlating with changes in circulatory markers. These data point to tissue-specific proinflammatory effects of high n-6 intake, but a net systemic anti-inflammatory effect as for n-3.
Collapse
Affiliation(s)
- Elise Grytten
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway; Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Johnny Laupsa-Borge
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway; Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway; Bevital AS, Bergen, Norway
| | - Kaya Cetin
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway; Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Pavol Bohov
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Jan Erik Nordrehaug
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Jon Skorve
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Rolf K Berge
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Elin Strand
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Bodil Bjørndal
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ottar K Nygård
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Espen Rostrup
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Gunnar Mellgren
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway; Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Simon N Dankel
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway; Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway.
| |
Collapse
|
11
|
Zhou L, Ran L, He Y, Huang Y. Mechanisms of microplastics on gastrointestinal injury and liver metabolism disorder (Review). Mol Med Rep 2025; 31:98. [PMID: 39981917 PMCID: PMC11865701 DOI: 10.3892/mmr.2025.13463] [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: 10/11/2024] [Accepted: 01/09/2025] [Indexed: 02/22/2025] Open
Abstract
With the high production and use of plastic products, a large amount of microplastics (MPs) is generated by degradation, which causes environmental pollution. MPs are particles with a diameter <5 mm; further degradation of MPs produces nano‑plastics (NPs), which could further increase the damage to cells when entering the human body. Therefore, the present review summarizes the effect of MP and NP deposition on the human gastrointestinal tract and the underlying injury mechanism of oxidative stress, inflammation and apoptosis, as well as the potential mechanism of glucose and liver lipid metabolism disorder. The present review provides a theoretical basis for research on the mechanisms of MPs in gastrointestinal injury and liver metabolism disorder. Further studies are needed for prevention and treatment of gastrointestinal diseases caused by MPs and NPs.
Collapse
Affiliation(s)
- Li Zhou
- Department of Gastroenterology and Hepatology, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing 400014, P.R. China
| | - Lidan Ran
- Department of Critical Care Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing 400014, P.R. China
| | - Yufen He
- Department of Gastroenterology and Hepatology, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing 400014, P.R. China
| | - Yaxi Huang
- Department of Gastroenterology and Hepatology, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing 400014, P.R. China
| |
Collapse
|
12
|
Jiang M, Huang J, Guo X, Fu W, Peng L, Wang Y, Liu W, Liu J, Zhou L, Xiao Y. HIF-3α/PPAR-γ Regulates Hypoxia Tolerance by Altering Glycolysis and Lipid Synthesis in Blunt Snout Bream ( Megalobrama amblycephala). Int J Mol Sci 2025; 26:2613. [PMID: 40141255 PMCID: PMC11942064 DOI: 10.3390/ijms26062613] [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] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Hypoxic stress causes cell damage and serious diseases in organisms, especially in aquatic animals. It is important to elucidate the changes in metabolic function caused by hypoxia and the mechanisms underlying these changes. This study focuses on the low oxygen tolerance feature of a new blunt snout bream strain (GBSBF1). Our data show that GBSBF1 has a different lipid and carbohydrate metabolism pattern than wild-type bream, with altering glycolysis and lipid synthesis. In GBSBF1, the expression levels of phd2 and vhl genes are significantly decreased, while the activation of HIF-3α protein is observed to have risen significantly. The results indicate that enhanced HIF-3α can positively regulate gpd1ab and gpam through PPAR-γ, which increases glucose metabolism and reduces lipolysis of GBSBF1. This research is beneficial for creating new aquaculture strains with low oxygen tolerance traits.
Collapse
Affiliation(s)
- Minggui Jiang
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| | - Jing Huang
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
| | - Xing Guo
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
| | - Wen Fu
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| | - Liangyue Peng
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| | - Yang Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (Y.W.); (L.Z.)
| | - Wenbin Liu
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| | - Jinhui Liu
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| | - Li Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (Y.W.); (L.Z.)
| | - Yamei Xiao
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| |
Collapse
|
13
|
Sun Y, Fan Z, Zhu X, Xia C, Shen G. Sodium butyrate activates peroxisome proliferator-activated receptor γ to suppress lithogenic diet-induced cholesterol gallstones in mice. Food Sci Biotechnol 2025; 34:1015-1026. [PMID: 39974854 PMCID: PMC11832876 DOI: 10.1007/s10068-024-01721-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 09/03/2024] [Accepted: 09/18/2024] [Indexed: 02/21/2025] Open
Abstract
Peroxisome proliferator-activated receptor γ (PPAR-γ) is crucial in forming cholesterol stones. Sodium butyrate (NaB), a short-chain fatty acid, shows potential for gallstone treatment by activating PPAR-γ. This study aimed to elucidate the effects of NaB on cholesterol gallstones in mice fed a lithogenic diet (LD). Ezetimibe (5 mg/day) was used as a positive control, and a PPAR-γ antagonist (CW9661, 4 mg/kg/day) was used to investigate PPAR-γ. Body weight, gallstone incidence, lipid concentrations in blood, bile, and liver, liver function evaluation, histological analysis, and cholesterol metabolism-related gene expression were evaluated. NaB and ezetimibe suppressed gallstone formation, serum AST, ALT, and ALP levels, and serum/liver TG and TC. They also reduced bile cholesterol and phospholipids, and liver histological damage. NaB activated PPAR-γ, CYP7A1, ABCA1, and ABCB11 while suppressing ABCG5/G8 gene expression. CW9661 reversed NaB's benefits in LD mice. This study provides scientific evidence that NaB activated PPAR-γ to improve gallstones. Supplementary Information The online version contains supplementary material available at 10.1007/s10068-024-01721-x.
Collapse
Affiliation(s)
- Yi Sun
- Department of General Surgery, Affiliated Xiaoshan Hospital Hangzhou Normal University, No.728 Yucai North Road, Xiaoshan District, Hangzhou, 311202 Zhejiang China
| | - Zhikun Fan
- Department of General Surgery, Affiliated Xiaoshan Hospital Hangzhou Normal University, No.728 Yucai North Road, Xiaoshan District, Hangzhou, 311202 Zhejiang China
| | - Xiaochao Zhu
- Department of General Surgery, Affiliated Xiaoshan Hospital Hangzhou Normal University, No.728 Yucai North Road, Xiaoshan District, Hangzhou, 311202 Zhejiang China
| | - Chao Xia
- Department of General Surgery, Affiliated Xiaoshan Hospital Hangzhou Normal University, No.728 Yucai North Road, Xiaoshan District, Hangzhou, 311202 Zhejiang China
| | - Guo Shen
- Department of General Surgery, Affiliated Xiaoshan Hospital Hangzhou Normal University, No.728 Yucai North Road, Xiaoshan District, Hangzhou, 311202 Zhejiang China
| |
Collapse
|
14
|
Zhang X, Wang SJ, Wan SC, Li X, Chen G. Ozone: complicated effects in central nervous system diseases. Med Gas Res 2025; 15:44-57. [PMID: 39436168 PMCID: PMC11515058 DOI: 10.4103/mgr.medgasres-d-24-00005] [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: 01/15/2024] [Revised: 06/20/2024] [Accepted: 08/25/2024] [Indexed: 10/23/2024] Open
Abstract
Oxidative stress is closely related to various diseases. Ozone can produce redox reactions through its unique response. As a source of the oxidative stress response, the strong oxidizing nature of ozone can cause severe damage to the body. On the other hand, low ozone concentrations can activate various mechanisms to combat oxidative stress and achieve therapeutic effects. Some animal experiments and clinical studies have revealed the potential medical value of ozone, indicating that ozone is not just a toxic gas. By reviewing the mechanism of ozone and its therapeutic value in treating central nervous system diseases (especially ischemic stroke and Alzheimer's disease) and the toxic effects of ozone, we find that ozone inhalation and a lack of antioxidants or excessive exposure lead to harmful impacts. However, with adequate antioxidants, ozone can transmit oxidative stress signals, reduce inflammation, reduce amyloid β peptide levels, and improve tissue oxygenation. Similar mechanisms to those of possible new drugs for treating ischemic stroke and Alzheimer's disease indicate the potential of ozone. Nevertheless, limited research has restricted the application of ozone. More studies are needed to reveal the exact dose-effect relationship and healing effect of ozone.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Shi-Jun Wang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Si-Cen Wan
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Xiang Li
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Gang Chen
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
15
|
Yu F, Chen J, Wang X, Hou S, Li H, Yao Y, He Y, Chen K. Metabolic reprogramming of peritoneal mesothelial cells in peritoneal dialysis-associated fibrosis: therapeutic targets and strategies. Cell Commun Signal 2025; 23:114. [PMID: 40016825 PMCID: PMC11866825 DOI: 10.1186/s12964-025-02113-2] [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] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 02/17/2025] [Indexed: 03/01/2025] Open
Abstract
Peritoneal dialysis (PD) is considered a life-saving treatment for end-stage renal disease. However, prolonged PD use can lead to the development of peritoneal fibrosis (PF), diminishing its efficacy. Peritoneal mesothelial cells (PMCs) are key initiators of PF when they become damaged. Exposure to high glucose‑based peritoneal dialysis fluids (PDFs) contributes to PF development by directly affecting highly metabolically active PMCs. Recent research indicates that PMCs undergo metabolic reprogramming when exposed to high-glucose PDFs, including enhanced glycolysis, impaired oxidative phosphorylation, abnormal lipid metabolism, and mitochondrial dysfunction. Although this metabolic transition temporarily compensates for the cellular damage and maintains energy levels, its long-term impact on peritoneal tissue is concerning. Multiple studies have identified a close association between this shift in energy metabolism and PF, and may promote the progression of PF through various molecular mechanisms. This review explores recent findings regarding the role and mechanism of PMC metabolic reprogramming in PF progression. Moreover, it provides a summary of potential therapeutic strategies aimed at various metabolic processes, including glucose metabolism, lipid metabolism, and mitochondrial function. The review establishes that targeting metabolic reprogramming in PMCs may be a novel strategy for preventing and treating PD-associated fibrosis.
Collapse
Affiliation(s)
- Fang Yu
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China
- Chongqing Key Laboratory of Precision Diagnosis and Treatment for Kidney Diseases, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China
| | - Jia Chen
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China
- Chongqing Key Laboratory of Precision Diagnosis and Treatment for Kidney Diseases, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China
| | - Xiaoyue Wang
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China
- Chongqing Key Laboratory of Precision Diagnosis and Treatment for Kidney Diseases, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China
| | - Shihui Hou
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China
| | - Hong Li
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China
| | - Yaru Yao
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China
| | - Yani He
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China.
- Chongqing Key Laboratory of Precision Diagnosis and Treatment for Kidney Diseases, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China.
- State Key Laboratory of Trauma and Chemical poisoning, Burns and Combined Injury, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China.
| | - Kehong Chen
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China.
- Chongqing Key Laboratory of Precision Diagnosis and Treatment for Kidney Diseases, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China.
- State Key Laboratory of Trauma and Chemical poisoning, Burns and Combined Injury, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China.
| |
Collapse
|
16
|
Merech F, Lara B, Rios D, Paparini D, Ramhorst R, Hauk V, Pérez Leirós C, Vota D. Vasoactive intestinal peptide induces metabolic rewiring of human-derived cytotrophoblast cells to promote cell migration. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119886. [PMID: 39653085 DOI: 10.1016/j.bbamcr.2024.119886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 11/28/2024] [Accepted: 12/02/2024] [Indexed: 12/16/2024]
Abstract
The placenta has an extraordinary metabolic rate with high oxygen consumption. Extravillous cytotrophoblast cells (EVT) metabolism and function are critical to sustain their invasive phenotype supporting fetal development. Deficient EVT function underlies pregnancy complications as preeclampsia (PE) and fetal growth restriction (FGR). The vasoactive intestinal peptide (VIP) promotes human cytotrophoblast cell migration and invasion through mTOR signaling pathways suggesting its crucial role during placentation. Here we explored fatty acid uptake as well as lipid and glucose metabolism in human-derived cytotrophoblast cell function upon VIP stimulation. We found that VIP induced long chain fatty acid (LCFAs) uptake along with the expression of FATP2 transporter, CPT1 fatty acid oxidation (FAO)-rate limiting step importer, and lipid droplet accumulation. VIP induced the expression of glucose 6-P-dehydrogenase, a rate-limiting enzyme of the pentose phosphate pathway (PPP) and pyruvate dehydrogenase complex enzyme DLAT E2, without altering lactate secretion. This metabolic rewiring of trophoblast cells induced by VIP takes place without compromising mitochondrial function or reactive oxygen species (ROS) production. Moreover, cytotrophoblast cell migration induced by VIP required the three glycolysis, oxidative phosphorylation (OXPHOS) and FAO pathways. Our results provide evidence supporting VIP as a metabolic regulatory peptide in cytotrophoblast cells sustaining proper placentation and fetal growth.
Collapse
Affiliation(s)
- Fátima Merech
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina
| | - Brenda Lara
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina
| | - Daiana Rios
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina
| | - Daniel Paparini
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina
| | - Rosanna Ramhorst
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina
| | - Vanesa Hauk
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina
| | - Claudia Pérez Leirós
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina.
| | - Daiana Vota
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina.
| |
Collapse
|
17
|
Chen F, Ma L, Liu Q, Zhou Z, Yi W. Recent advances and therapeutic applications of PPARγ-targeted ligands based on the inhibition mechanism of Ser273 phosphorylation. Metabolism 2025; 163:156097. [PMID: 39637972 DOI: 10.1016/j.metabol.2024.156097] [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: 09/30/2024] [Revised: 11/27/2024] [Accepted: 11/30/2024] [Indexed: 12/07/2024]
Abstract
PPARγ functions as a master ligand-dependent transcription factor that regulates the expressions of a variety of key genes related to metabolic homeostasis and inflammatory immunity. It has been recognized as a popular and druggable target in modern drug discovery. Similar to other nuclear receptors, PPARγ is a phosphoprotein, and its biological functions are regulated by phosphorylation, especially at Ser273 site which is mediated by CDK5 or ERK. In the past decade, the excessive level of PPARγ-Ser273 phosphorylation has been confirmed to be a crucial factor in promoting the occurrence and development of some major diseases. Ligands capable of inhibiting PPARγ-Ser273 phosphorylation have shown great potentials for treatment. Despite these achievements, to our knowledge, no related review focusing on this topic has been conducted so far. Therefore, we herein summarize the basic knowledge of PPARγ and CDK5/ERK-mediated PPARγ-Ser273 phosphorylation as well as its physiopathological role in representative diseases. We also review the developments and therapeutic applications of PPARγ-targeted ligands based on this mechanism. Finally, we suggest several directions for future investigations. We expect that this review can evoke more inspiration of scientific communities, ultimately facilitating the promotion of the PPARγ-Ser273 phosphorylation-involved mechanism as a promising breakthrough point for addressing the clinical treatment of human diseases.
Collapse
Affiliation(s)
- Fangyuan Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation & Molecular Target and Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Lei Ma
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation & Molecular Target and Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Qingmei Liu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation & Molecular Target and Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Zhi Zhou
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation & Molecular Target and Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China.
| | - Wei Yi
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation & Molecular Target and Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China.
| |
Collapse
|
18
|
Collodel G, Moretti E, Marcucci C, Liguori L, Marchini D, Corsaro R, Centini G, Signorini C. PPARγ Expression in Human Spermatozoa and Its Relationship with Seminal F 2-Isoprostanes and Resolvin D1 in the Presence of Varicocele and Urogenital Infections. BIOLOGY 2025; 14:137. [PMID: 40001905 PMCID: PMC11851876 DOI: 10.3390/biology14020137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025]
Abstract
Peroxisome Proliferator-Activated Receptor Gamma (PPARγ) is a regulating agent in antioxidant response also involved in controlling inflammation. The impact of varicocele and urogenital infections on sperm PPARγ expression was studied. The PPARγ gene expression was investigated in spermatozoa of 26 normozoospermic men grouped according to their clinical conditions: normal semen parameters (N), normal semen parameters and varicocele (N + V), and normal semen parameters and urogenital infections (N + UI). Sperm PPARγ expression was correlated with F2-isoprostanes (F2-IsoPs), as markers of lipid peroxidation, and Resolvin D1 (RvD1), a pro-resolving mediator in inflammation. Sperm PPARγ expression was evaluated through comparative real-time PCR, and F2-IsoPs and RvD1 were quantified in the seminal plasma via GC/NCI-MS/MS and immunoassay, respectively. PPARγ expression correlates positively with sperm morphology and vitality and negatively with F2-IsoPs and RvD1. Sperm morphology positively correlates with vitality and negatively with F2-IsoP and RvD1 levels. Despite the normozoospermia in the three examined groups, sperm morphology and PPARγ expression were significantly reduced in N + V and N + UI groups compared to the N group. Additionally, F2-IsoP and RvD1 levels were elevated in N + V and N + UI patients. These data suggest that PPARγ expression is compromised by inflammation and lipoperoxidation, providing new insights to further explore new possibilities of targeted treatment of male infertility.
Collapse
Affiliation(s)
- Giulia Collodel
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (G.C.); (C.M.); (L.L.); (R.C.); (G.C.); (C.S.)
| | - Elena Moretti
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (G.C.); (C.M.); (L.L.); (R.C.); (G.C.); (C.S.)
| | - Caterina Marcucci
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (G.C.); (C.M.); (L.L.); (R.C.); (G.C.); (C.S.)
| | - Laura Liguori
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (G.C.); (C.M.); (L.L.); (R.C.); (G.C.); (C.S.)
| | - Daniela Marchini
- Department of Life Sciences, University of Siena, 53100 Siena, Italy;
| | - Roberta Corsaro
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (G.C.); (C.M.); (L.L.); (R.C.); (G.C.); (C.S.)
| | - Gabriele Centini
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (G.C.); (C.M.); (L.L.); (R.C.); (G.C.); (C.S.)
| | - Cinzia Signorini
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (G.C.); (C.M.); (L.L.); (R.C.); (G.C.); (C.S.)
| |
Collapse
|
19
|
Zhang C, Fang Y, Guo M, Tang L, Xing Y, Zhou J, Guo Y, Gu Y, Wen Q, Gao N, Xu H, Qiao H. Q11, a CYP2E1 inhibitor, exerts anti-hepatocellular carcinoma effect by inhibiting M2 macrophage polarization. Cancer Immunol Immunother 2024; 74:35. [PMID: 39738913 PMCID: PMC11685367 DOI: 10.1007/s00262-024-03912-1] [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: 10/20/2024] [Accepted: 12/01/2024] [Indexed: 01/02/2025]
Abstract
Despite significant advancements in cancer immunotherapy, many patients continue to respond poorly. Novel therapeutic strategies and drugs are urgently needed. Here, we found that CYP2E1 is upregulated in M2 macrophages. The CYP2E1 inhibitor, Q11, could inhibit M2 macrophage polarization, while CYP2E1 overexpression could promote it. Increased levels of CYP2E1 and M2 macrophages in the tumor microenvironment of HCC patients correlate with poor prognosis. Q11 could inhibit tumor cells by targeting M2 macrophages rather than directly attacking tumor cells. Both Q11 and Cyp2e1 knockout could effectively suppress tumor growth. Q11 reduces the production of CYP2E1 metabolites ( ±)9(10)-DiHOME and ( ±)12(13)-DiHOME, thus attenuating PPARγ activation and M2 macrophage polarization. In summary, our findings suggest that Q11 could suppress M2 macrophage polarization by modulating the CYP2E1/( ±)9(10)-DiHOME or ( ±)12(13)-DiHOME/PPARγ axis, indicating that CYP2E1 may be a potential therapeutic target for HCC, and its inhibitor Q11 may be a potential drug for the treatment of HCC.
Collapse
Affiliation(s)
- Cunzhen Zhang
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, 450001, China
| | - Yan Fang
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, 450001, China
| | - Mengxue Guo
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, 450001, China
| | - Liming Tang
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, 450001, China
| | - Yurong Xing
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China
| | - Jun Zhou
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, 450001, China
| | - Yuanyuan Guo
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, 450001, China
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China
| | - Yuhan Gu
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, 450001, China
| | - Qiang Wen
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, 450001, China
| | - Na Gao
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, 450001, China
| | - Haiwei Xu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Hailing Qiao
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
20
|
Li Q, Zhang Q, Su S, Yang S, Shao J, Guan W, Zhang S. Maternal fish oil supplementation enhances nutrient transport in the placenta and milk biosynthesis in the mammary gland via the GPR120 signaling pathway. J Adv Res 2024:S2090-1232(24)00607-6. [PMID: 39706333 DOI: 10.1016/j.jare.2024.12.029] [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: 11/14/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024] Open
Abstract
INTRODUCTION Maternal fish oil (FO) supplementation during pregnancy has been shown to improve pregnancy outcomes. FO is recognized as dietary source for n-3 polyunsaturated fatty acids (n-3 PUFAs). While early research has focused on the benefits of n-3 PUFAs for fetal neurodevelopment, retinal maturation and neonatal behavior, their roles in the placenta during late pregnancy and in the mammary gland during lactation still remain unknow. OBJECTIVES Here, we aim to clarify the mechanisms by which maternal supplementation with FO during pregnancy and lactation affects placental and mammary gland function. METHODS We evaluated the effects of FO on maternal placental nutrient transport, mammary gland milk synthesis and offspring growth. We then explored the molecular mechanisms by which docosahexaenoic acid (DHA) affects the biological function of placental trophoblast cells and mammary epithelial cells through in vitro experiments. Finally, a lipopolysaccharide-challenged experiment was performed to access the potential of maternal FO supplementation in alleviating offspring intestinal inflammation. RESULTS Maternal supplementation with FO during late pregnancy increased offspring birth weight, associated with enhanced maternal placental vascularization and nutrient transporter abundance. Additionally, maternal FO supplementation during lactation improved milk biosynthesis, increasing the fat, protein, and non-fat solids content in both colostrum and mature milk, thereby promoting offspring growth. The stimulatory effects of DHA on nutrient transportation in placental trophoblast cells and nutrient secretion in mammary gland epithelial cells were mediated by GPR120 signaling pathways. Furthermore, maternal FO supplementation strengthened the placental barrier, reduced placental inflammation, oxidative stress and alleviated lipopolysaccharide-induced intestinal inflammation in offspring. CONCLUSION Maternal FO supplementation during late pregnancy and lactation enhances offspring growth by increasing placental nutrient transport and milk biosynthesis, mediated by GPR120. Additionally, maternal FO supplementation reduces the susceptibility of offspring to intestinal inflammation.
Collapse
Affiliation(s)
- Qihui Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | - Qianzi Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Senlin Su
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Siwang Yang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jiayuan Shao
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Wutai Guan
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Shihai Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
21
|
Mérida DM, Rey-García J, Moreno-Franco B, Guallar-Castillón P. Acrylamide Exposure and Cardiovascular Risk: A Systematic Review. Nutrients 2024; 16:4279. [PMID: 39770901 PMCID: PMC11677207 DOI: 10.3390/nu16244279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 01/30/2025] Open
Abstract
Background/Objectives: Acrylamide is a food contaminant formed during high-temperature cooking processes, leading to unintentional human exposure. Diet is the primary source for non-smokers, with potatoes, cereals, and coffee being the main contributors. While animal studies have demonstrated that acrylamide is neurotoxic, genotoxic, mutagenic, and cardiotoxic, its effects on human cardiovascular health remain poorly understood. This study aimed to evaluate the association between acrylamide exposure and cardiovascular risk. Methods: A comprehensive literature search was conducted across four databases without restrictions on publication year or language (last search: 1 July 2024). The risk of bias was assessed using the Joanna Briggs Institute critical appraisal tools. Results: In total, 28 studies were included, predominantly from the US NHANES sample and with cross-sectional designs. Higher acrylamide exposure was associated with an increased risk of cardiovascular mortality but was inversely associated with glucose and lipid levels, as well as key cardiovascular risk factors such as diabetes, obesity, and metabolic syndrome. Conversely, glycidamide-acrylamide's most reactive metabolite-was positively associated with elevated glucose and lipid levels, higher systolic blood pressure, and increased obesity prevalence. Conclusions: These findings suggest that the adverse cardiovascular effects of acrylamide may be mediated by its conversion to glycidamide. Further research is necessary to fully elucidate the impact of acrylamide on cardiovascular health. Meanwhile, public health efforts should continue to focus on mitigation strategies within the food industry and raising public awareness about exposure.
Collapse
Affiliation(s)
- Diana María Mérida
- Department of Preventive Medicine and Public Health, School of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain
- Department of Pharmacoepidemiology and Biostatistics, Fundación Teófilo Hernando, 28290 Las Rozas de Madrid, Spain
- CIBERESP (CIBER of Epidemiology and Public Health), 28029 Madrid, Spain
| | - Jimena Rey-García
- Department of Internal Medicine, Hospital Universitario Rey Juan Carlos, IIS-FJD, 28933 Móstoles, Spain
| | - Belén Moreno-Franco
- Instituto de Investigación Sanitaria Aragón, Hospital Universitario Miguel Servet, 50009 Zaragoza, Spain
- Department of Preventive Medicine and Public Health, Universidad de Zaragoza, 50009 Zaragoza, Spain
- CIBERCV (CIBER of Cardiovascular Diseases), 28029 Madrid, Spain
| | - Pilar Guallar-Castillón
- Department of Preventive Medicine and Public Health, School of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain
- CIBERESP (CIBER of Epidemiology and Public Health), 28029 Madrid, Spain
- IMDEA-Food Institute, CEI UAM+CSIC, Carretera de Cantoblanco 8, 28049 Madrid, Spain
| |
Collapse
|
22
|
Yang X, Li J, Xu C, Zhang G, Che X, Yang J. Potential mechanisms of rheumatoid arthritis therapy: Focus on macrophage polarization. Int Immunopharmacol 2024; 142:113058. [PMID: 39236455 DOI: 10.1016/j.intimp.2024.113058] [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: 07/18/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/07/2024]
Abstract
Rheumatoid arthritis (RA) is an autoimmune inflammatory disease that affects multiple organs and systems in the human body, often leading to disability. Its pathogenesis is complex, and the long-term use of traditional anti-rheumatic drugs frequently results in severe toxic side effects. Therefore, the search for a safer and more effective antirheumatic drug is extremely important for the treatment of RA. As important immune cells in the body, macrophages are polarized. Under pathological conditions, macrophages undergo proliferation and are recruited to diseased tissues upon stimulation. In the local microenvironment, they polarize into different types of macrophages in response to specific factors and perform unique functions and roles. Previous studies have shown that there is a link between macrophage polarization and RA, indicating that certain active ingredients can ameliorate RA symptoms through macrophage polarization. Notably, Traditional Chinese medicine (TCM) monomer component and compounds demonstrate a particular advantage in this process. Building upon this insight, we reviewed and analyzed recent studies to offer valuable and meaningful insights and directions for the development and application of anti-rheumatic drugs.
Collapse
Affiliation(s)
- Xinyu Yang
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinling Li
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chengchao Xu
- College of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guangheng Zhang
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xinzhen Che
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiguo Yang
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
23
|
Li L, Zhao S, Leng Z, Chen S, Shi Y, Shi L, Li J, Mao K, Tang H, Meng B, Wang Y, Shang G, Liu H. Pathological mechanisms and related markers of steroid-induced osteonecrosis of the femoral head. Ann Med 2024; 56:2416070. [PMID: 39529511 PMCID: PMC11559024 DOI: 10.1080/07853890.2024.2416070] [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: 04/05/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Osteonecrosis of the femoral head (ONFH) is a refractory orthopedic disease with a high disability rate. Long-term administration of steroids is the most common pathogenic factor for non-traumatic ONFH. Early diagnosis of steroid-induced osteonecrosis of the femoral head (SONFH) is difficult and mainly depends on imaging. OBJECTIVES The objectives of this review were to examine the pathological mechanisms of SONFH, summarize related markers of SONFH, and identify areas for future studies. METHODS We reviewed studies on pathological mechanisms and related markers of SONFH and discussed the relationship between them, as well as clinical applications and the outlook of potential markers. RESULTS The pathological mechanisms of SONFH included decreased osteogenesis, lipid accumulation, increased intraosseous pressure, and microcirculation disruption. Differential proteomics and genomics play crucial roles in the occurrence, progression, and outcome of SONFH, providing novel insights into SONFH. Additionally, the biological functions of mesenchymal stem cells (MSCs) and exosomes (Exos) in SONFH have attracted increasing attention. CONCLUSIONS The pathological mechanisms of SONFH are complex. The related markers mentioned in the current review can predict the occurrence and progression of SONFH, which will help provide effective early clinical prevention and treatment strategies for SONFH.
Collapse
Affiliation(s)
- Longyu Li
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shangkun Zhao
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zikuan Leng
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Songfeng Chen
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yifang Shi
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lijun Shi
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinfeng Li
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Keya Mao
- Department of Orthopedics, The Chinese PLA General Hospital, Beijing, China
| | - Hai Tang
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Bin Meng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yisheng Wang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guowei Shang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongjian Liu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
24
|
Wang C, Zhao J, Feng X, Zhao W, Ma R, Yu B, Xue L, Wang H, Chen Y, Zhang J, Gu Y. bta-miR-224 regulates milk fat metabolism by targeting FABP4 in bovine mammary epithelial cells. Genomics 2024; 116:110955. [PMID: 39481579 DOI: 10.1016/j.ygeno.2024.110955] [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: 08/13/2024] [Revised: 10/13/2024] [Accepted: 10/27/2024] [Indexed: 11/02/2024]
Abstract
Milk fat is produced and secreted by the mammary gland, which is mainly regulated by diet and gene-molecule network. Therefore, understanding the molecular mechanism of milk fat synthesis is of practical significance for improving milk quality. Fatty acid-binding protein 4 (FABP4) is a candidate messenger RNA (mRNA) closely linked to milk fat metabolism obtained from transcriptomic analysis of mammary epithelial cells of cows in the pre-existing high- and low-milk-fat groups, and its expression pattern and function are still unclear. The qRT-PCR results depicted that FABP4 was highly expressed in bovine mammary epithelial cells (BMECs) in breast tissues and the high milk fat group. Subsequently, the regulatory effects of FABP4 on BMECs were analyzed by CCK8, EdU, and flow cytometry, and the results demonstrated that FABP4 inhibited the proliferation and viability of BMECs and promoted their apoptosis. Next, the effect of FABP4 on milk lipid metabolism was explored. pEGFP-N1-FABP4 was transfected into BMECs, and FABP4 upregulated the expression levels of the milk lipid marker genes XDH, PPARG, and ACSS2, and promoted the formation of triglycerides (TGs), cholesterol, lipid droplets, and β-casein. Strong interactions between FABP4 and PPARG were identified using STRING prediction. Western blotting revealed that FABP4 interacted with PPARG to promote PPARG expression, while the opposite result was observed after interfering with FABP4. The gene regulation of microRNA (miRNA) is essential for fatty acid metabolism and synthesis. Predicted by website and combined with pre-miRNA transcriptome sequencing results, we hypothesized that FABP4 might be the target gene of bta-miR-224. The results of the dual-luciferase reporter gene and qRT-PCR revealed that bta-miR-224 negatively regulated FABP4 expression by targeting the 3'-UTR of FABP4. By exploring the function of bta-miR-224, we observed that bta-miR-224 mimics downregulated the expression of the milk fat marker genes AGPAT6, ACSS2, and XDH and inhibited TG synthesis and lipid droplet secretion. However, the bta-miR-224 inhibitor depicted the opposite results. In conclusion, FABP4 plays a crucial role in regulating BMEC proliferation and differentiation. Bta-miR-224 targeting FABP4 may promote biological processes such as TG synthesis and lipid droplet formation through PPARG, which lays a solid foundation for further analysis of the functional mechanism of milk lipid metabolism in dairy cows from a miRNA-mRNA perspective.
Collapse
Affiliation(s)
- Chuanchuan Wang
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding in Ningxia, Ningxia University, Yinchuan 750021, China
| | - Jinyan Zhao
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding in Ningxia, Ningxia University, Yinchuan 750021, China
| | - Xiaofang Feng
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding in Ningxia, Ningxia University, Yinchuan 750021, China
| | - Wei Zhao
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding in Ningxia, Ningxia University, Yinchuan 750021, China
| | - Ruoshuang Ma
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding in Ningxia, Ningxia University, Yinchuan 750021, China
| | - Baojun Yu
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding in Ningxia, Ningxia University, Yinchuan 750021, China
| | - Lin Xue
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding in Ningxia, Ningxia University, Yinchuan 750021, China
| | - Hua Wang
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding in Ningxia, Ningxia University, Yinchuan 750021, China
| | - Yafei Chen
- Yinchuan Animal Husbandry Technical Extension and Service Centre, Yinchuan 750021, China
| | - Juan Zhang
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding in Ningxia, Ningxia University, Yinchuan 750021, China
| | - Yaling Gu
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding in Ningxia, Ningxia University, Yinchuan 750021, China.
| |
Collapse
|
25
|
D P, Hani U, Haider N, Talath S, Shanmugarajan D, P P, P A, Prashantha Kumar BR. Novel PPAR-γ agonists as potential neuroprotective agents against Alzheimer's disease: rational design, synthesis , in silico evaluation, PPAR-γ binding assay and transactivation and expression studies. RSC Adv 2024; 14:33247-33266. [PMID: 39434987 PMCID: PMC11492828 DOI: 10.1039/d4ra06330a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 09/30/2024] [Indexed: 10/23/2024] Open
Abstract
Alzheimer's disease (AD) is a neurological disorder. It is caused by accumulation of amyloid beta (Aβ) plaques and tau tangles, which gradually leads to cognitive decline and memory loss. Peroxisome proliferator-activated receptor gamma (PPAR-γ), a nuclear receptor, plays a significant role in regulating genes responsible for metabolism and inflammation. Studies have shown that PPAR-γ activation has neuroprotective effects, can potentially reduce inflammation and oxidative stress, and stimulates mitochondrial biogenesis. Current study presents the design, synthesis and in vitro evaluation of PPAR-γ agonists for AD that are tailored to optimize binding with the PPAR-γ receptor. The compounds 4a, 4h and 4j exhibited notable binding affinities towards PPAR-γ LBD, with IC50 values of 8.607, 9.242, and 5.974 μM, respectively, in TR-FRET binding assay. These compounds were cell proliferative and non-cytotoxic in a neuroblastoma cell line (SH-SY5Y). They also demonstrated dose-dependent PPAR-γ activation in transactivation assay. Their neuroprotective effect was studied based on their anti-inflammatory and anti-oxidant potential by reducing the levels of proinflammatory markers (TNF-α, IL-6 and IL-1β) and ROS in Aβ-induced SH-SY5Y neuroblastoma cells using a flow cytometry method. The synthesized compounds also showed interactions in molecular docking study with the PPAR-γ receptor and demonstrated good stability in MD simulation. Our results highlight that through activation of PPAR-γ, the compounds 4a, 4h and 4j offer neuroprotective effects by reducing neuroinflammation and oxidative stress, and hence, they may be considered lead molecules for treating AD.
Collapse
Affiliation(s)
- Priya D
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research Sri Shivarathreeshwara Nagara Mysuru 570015 India +91-821-2548359 +91-821-2548353
| | - Umme Hani
- Department of Pharmaceutics, College of Pharmacy, King Khalid University Abha 62529 Saudi Arabia
| | - Nazima Haider
- Department of Pathology, College of Medicine, King Khalid University Abha 62529 Saudi Arabia
| | - Sirajunisa Talath
- Department of Pharmaceutical Chemistry, RAK College of Pharmacy, RAK Medical and Health Sciences University Ras Al Khaimah 11172 United Arab Emirates
| | - Dhivya Shanmugarajan
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research Sri Shivarathreeshwara Nagara Mysuru 570015 India +91-821-2548359 +91-821-2548353
| | - Prabitha P
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research Sri Shivarathreeshwara Nagara Mysuru 570015 India +91-821-2548359 +91-821-2548353
| | - Archana P
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research Sri Shivarathreeshwara Nagara Mysuru 570015 India +91-821-2548359 +91-821-2548353
| | - B R Prashantha Kumar
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research Sri Shivarathreeshwara Nagara Mysuru 570015 India +91-821-2548359 +91-821-2548353
| |
Collapse
|
26
|
Lima GB, Figueiredo N, Kattah FM, Oliveira ES, Horst MA, Dâmaso AR, Oyama LM, Whitton RGM, de Souza GIMH, Lima GC, Mota JF, Campos RMS, Corgosinho FC. Serum Fatty Acids and Inflammatory Patterns in Severe Obesity: A Preliminary Investigation in Women. Biomedicines 2024; 12:2248. [PMID: 39457561 PMCID: PMC11505423 DOI: 10.3390/biomedicines12102248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/29/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Inflammation plays a central role in many chronic diseases that characterize modern society. Leptin/adiponectin and adiponectin/leptin ratios have been recognized as notable markers of dysfunctional adipose tissue and, consequently, an inflammatory state. Methods: Blood samples were collected from 41 adult volunteers (40.2 ± 8.3 years) diagnosed with severe obesity (BMI 46.99; 42.98-51.91 kg/m2). The adipokines were quantified using an enzyme-linked immunosorbent assay, while the serum fatty acid analysis was conducted using chromatography. Results: The results unveiled a positive correlation between the leptin/adiponectin ratio and the 20:3n6 fatty acid (r = 0.52, p = 0.001), as well as a similar positive correlation between the adiponectin/leptin ratio and the 22:6n3 fatty acid (r = 0.74, p = 0.001). In the regression analysis, the 22:6n3 fatty acid predicted the adiponectin/leptin ratio (β = 0.76, p < 0.001), whereas C20:3 n-6 was a predictor for inflammatory markers (β = 4.84, p < 0.001). Conclusions: In conclusion, the 22:6n3 fatty acid was demonstrated to be a predictive factor for the adiponectin/leptin ratio and C20:3 n-6 was a predictor for inflammatory markers. This discovery, novel within this population, can help develop new intervention strategies aimed at controlling the inflammatory status in individuals classified as having severe obesity.
Collapse
Affiliation(s)
- Gislene B. Lima
- Postgraduate Program in Nutrition and Health, Faculty of Nutrition, Federal University of Goiás (UFG), Goiânia 74690-900, Brazil; (F.M.K.); (E.S.O.); (M.A.H.); (G.C.L.); (J.F.M.); (F.C.C.)
| | - Nayra Figueiredo
- Postgraduate Program in Health Sciences, Faculty of Medicine, Federal University of Goiás (UFG), Goiânia 74690-900, Brazil;
| | - Fabiana M. Kattah
- Postgraduate Program in Nutrition and Health, Faculty of Nutrition, Federal University of Goiás (UFG), Goiânia 74690-900, Brazil; (F.M.K.); (E.S.O.); (M.A.H.); (G.C.L.); (J.F.M.); (F.C.C.)
| | - Emilly S. Oliveira
- Postgraduate Program in Nutrition and Health, Faculty of Nutrition, Federal University of Goiás (UFG), Goiânia 74690-900, Brazil; (F.M.K.); (E.S.O.); (M.A.H.); (G.C.L.); (J.F.M.); (F.C.C.)
| | - Maria A. Horst
- Postgraduate Program in Nutrition and Health, Faculty of Nutrition, Federal University of Goiás (UFG), Goiânia 74690-900, Brazil; (F.M.K.); (E.S.O.); (M.A.H.); (G.C.L.); (J.F.M.); (F.C.C.)
| | - Ana R. Dâmaso
- Interdisciplinary Postgraduate Program in Health Sciences, Federal University of São Paulo (UNIFESP), São Paulo 04039-032, Brazil; (A.R.D.); (L.M.O.); (G.I.M.H.d.S.); (R.M.S.C.)
| | - Lila M. Oyama
- Interdisciplinary Postgraduate Program in Health Sciences, Federal University of São Paulo (UNIFESP), São Paulo 04039-032, Brazil; (A.R.D.); (L.M.O.); (G.I.M.H.d.S.); (R.M.S.C.)
| | - Renata G. M. Whitton
- Institute of Biosciences, Federal University of São Paulo (USP), São Paulo 05508-900, Brazil;
| | - Gabriel I. M. H. de Souza
- Interdisciplinary Postgraduate Program in Health Sciences, Federal University of São Paulo (UNIFESP), São Paulo 04039-032, Brazil; (A.R.D.); (L.M.O.); (G.I.M.H.d.S.); (R.M.S.C.)
| | - Glaucia C. Lima
- Postgraduate Program in Nutrition and Health, Faculty of Nutrition, Federal University of Goiás (UFG), Goiânia 74690-900, Brazil; (F.M.K.); (E.S.O.); (M.A.H.); (G.C.L.); (J.F.M.); (F.C.C.)
| | - João F. Mota
- Postgraduate Program in Nutrition and Health, Faculty of Nutrition, Federal University of Goiás (UFG), Goiânia 74690-900, Brazil; (F.M.K.); (E.S.O.); (M.A.H.); (G.C.L.); (J.F.M.); (F.C.C.)
- Postgraduate Program in Health Sciences, Faculty of Medicine, Federal University of Goiás (UFG), Goiânia 74690-900, Brazil;
| | - Raquel M. S. Campos
- Interdisciplinary Postgraduate Program in Health Sciences, Federal University of São Paulo (UNIFESP), São Paulo 04039-032, Brazil; (A.R.D.); (L.M.O.); (G.I.M.H.d.S.); (R.M.S.C.)
| | - Flávia C. Corgosinho
- Postgraduate Program in Nutrition and Health, Faculty of Nutrition, Federal University of Goiás (UFG), Goiânia 74690-900, Brazil; (F.M.K.); (E.S.O.); (M.A.H.); (G.C.L.); (J.F.M.); (F.C.C.)
- Postgraduate Program in Health Sciences, Faculty of Medicine, Federal University of Goiás (UFG), Goiânia 74690-900, Brazil;
| |
Collapse
|
27
|
Li K, Yan J, Wang S, Zhu C, Zhu Q, Lu S, Hu P, Dessie T, Kim IH, Ahmed AA, Liu HY, Ennab W, Cai D. Transcriptome analysis provides new insights into the response of canine intestinal epithelial cells treated by sulforaphane: a natural product of cruciferous origin. Front Vet Sci 2024; 11:1460500. [PMID: 39415954 PMCID: PMC11479859 DOI: 10.3389/fvets.2024.1460500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 09/12/2024] [Indexed: 10/19/2024] Open
Abstract
This study presents a comprehensive transcriptome analysis of canine intestinal epithelial cells following treatment with sulforaphane (SFN), a naturally occurring compound found in cruciferous vegetables with established anti-inflammatory and antioxidant properties. Through high-throughput sequencing, we identified 29,993 genes, among which 1,612 were differentially expressed, with 792 up-regulated and 820 down-regulated in response to SFN treatment. Our analysis revealed significant enrichment of genes in pathways associated with the inflammatory response, lipid metabolism, oxidative stress response, and T-cell mediated immunity, suggesting SFN's potential in modulating these biological processes. Notably, the PPARγ gene, which plays a crucial role in the body's oxidative stress and inflammatory response, was highly up-regulated, indicating its possible centrality in SFN's effects. Gene-gene interaction analysis further supported SFN's role in alleviating inflammation through PPARγ, with key genes in oxidative stress and inflammatory response pathways showing significant correlations with PPARγ. Overall, our findings provide molecular evidence for SFN's protective effects on canine intestinal health, potentially through the modulation of inflammatory and oxidative stress pathways, with PPARγ emerging as a critical mediator.
Collapse
Affiliation(s)
- Kaiqi Li
- Laboratory of Animal Physiology and Molecular Nutrition, Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Jin Yan
- Laboratory of Animal Physiology and Molecular Nutrition, Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Shiqi Wang
- Laboratory of Animal Physiology and Molecular Nutrition, Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Chuyang Zhu
- Laboratory of Animal Physiology and Molecular Nutrition, Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Qi Zhu
- Laboratory of Animal Physiology and Molecular Nutrition, Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Sichen Lu
- Laboratory of Animal Physiology and Molecular Nutrition, Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Ping Hu
- Laboratory of Animal Physiology and Molecular Nutrition, Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Tadelle Dessie
- International Livestock Research Institute, Addis Ababa, Ethiopia
| | - In Ho Kim
- Department of Animal Resource and Science, Dankook University, Cheonan, Republic of Korea
| | - Abdelkareem A. Ahmed
- Department of Veterinary Biomedical Sciences, Botswana University of Agriculture and Agriculture and Natural Resources, Gaborone, Botswana
| | - Hao-Yu Liu
- Laboratory of Animal Physiology and Molecular Nutrition, Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Wael Ennab
- Laboratory of Animal Physiology and Molecular Nutrition, Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Demin Cai
- Laboratory of Animal Physiology and Molecular Nutrition, Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou, Jiangsu, China
| |
Collapse
|
28
|
Tian R, Miao L, Cheang WS. Effects of Pterostilbene on Cardiovascular Health and Disease. Curr Issues Mol Biol 2024; 46:9576-9587. [PMID: 39329921 PMCID: PMC11430207 DOI: 10.3390/cimb46090569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 08/25/2024] [Accepted: 08/27/2024] [Indexed: 09/28/2024] Open
Abstract
Pterostilbene is a phenolic compound commonly found in blueberries, peanuts, grapes, and other plants. It is a dimethoxy derivative of resveratrol. In recent years, it has gained significant attention due to its remarkable anti-inflammatory and antioxidant effects. In addition, its high bioavailability and low toxicity in many species has contributed to its promising research prospects. Cardiovascular disease is closely related to pathological processes such as inflammation and oxidative stress, which aligns well with the treatment applications of pterostilbene. As a result, numerous studies have investigated the effects of pterostilbene on cardiovascular health and disease. This paper summarizes the current research on pterostilbene, with a specific focus on its potential therapeutic role in treating cardiovascular disease.
Collapse
Affiliation(s)
- Rui Tian
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Lingchao Miao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Wai-San Cheang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| |
Collapse
|
29
|
Gao R, Zhu Q, Huang L, Fan X, Teng X, Miao Y. LEP Gene Promotes Milk Fat Synthesis via the JAK2-STAT3 and mTOR Signaling Pathways in Buffalo Mammary Epithelial Cells. Animals (Basel) 2024; 14:2446. [PMID: 39199979 PMCID: PMC11350831 DOI: 10.3390/ani14162446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/16/2024] [Accepted: 08/21/2024] [Indexed: 09/01/2024] Open
Abstract
Leptin (LEP), a protein hormone well-known for its role in metabolic regulation, has recently been linked to lipid metabolism in cattle. However, its function in buffalo mammary glands remains unclear. To address this issue, we isolated and identified the LEP gene and conducted experiments to investigate its function in buffalo mammary epithelial cells (BuMECs). In this study, two transcript variants of LEP, designated as LEP_X1 and LEP_X2, were identified. The coding sequences (CDS) of LEP_X1 and LEP_X2 are 504 bp and 579 bp in length, encoding 167 and 192 amino acid residues, respectively. Bioinformatics analysis revealed that LEP_X2 is a hydrophobic protein with an isoelectric point below 7 and contains a signal peptide, while LEP_X1 is hydrophilic and lacks a signal peptide. Our study found that LEP gene expression in lactating BuMECs was significantly higher than in non-lactating cells, with LEP_X2 expression remarkably higher than LEP_X1 in lactating BuMECs. Overexpression of both LEP_X1 and LEP_X2 significantly promoted the expression of genes related to milk fat synthesis in lactating BuMECs, including STAT3, PI3K, mTOR, SCD, and SREBF1, accompanied by an increase in cellular triglycerides (TG). Interestingly, LEP_X2 overexpression significantly suppressed LEP_X1 expression while increasing intracellular TG concentration by 12.10-fold compared to LEP_X1 overexpression, suggesting an antagonistic relationship between the two variants and supposing LEP_X2 plays a dominant role in milk fat synthesis in lactating BuMECs. Additionally, four nucleotide substitutions were identified in the buffalo LEP CDS, including a nonsynonymous substitution c.148C>T (p.Arg50Cys), which was predicted to decrease the stability of the LEP protein without affecting its function. These results collectively underscore the significant role of LEP in milk fat synthesis and can provide a basis for molecular breeding strategies of buffalo.
Collapse
Affiliation(s)
| | | | | | | | | | - Yongwang Miao
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (R.G.); (Q.Z.); (L.H.); (X.F.); (X.T.)
| |
Collapse
|
30
|
Dowker-Key PD, Jadi PK, Gill NB, Hubbard KN, Elshaarrawi A, Alfatlawy ND, Bettaieb A. A Closer Look into White Adipose Tissue Biology and the Molecular Regulation of Stem Cell Commitment and Differentiation. Genes (Basel) 2024; 15:1017. [PMID: 39202377 PMCID: PMC11353785 DOI: 10.3390/genes15081017] [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: 06/24/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 09/03/2024] Open
Abstract
White adipose tissue (WAT) makes up about 20-25% of total body mass in healthy individuals and is crucial for regulating various metabolic processes, including energy metabolism, endocrine function, immunity, and reproduction. In adipose tissue research, "adipogenesis" is commonly used to refer to the process of adipocyte formation, spanning from stem cell commitment to the development of mature, functional adipocytes. Although, this term should encompass a wide range of processes beyond commitment and differentiation, to also include other stages of adipose tissue development such as hypertrophy, hyperplasia, angiogenesis, macrophage infiltration, polarization, etc.… collectively, referred to herein as the adipogenic cycle. The term "differentiation", conversely, should only be used to refer to the process by which committed stem cells progress through distinct phases of subsequent differentiation. Recognizing this distinction is essential for accurately interpreting research findings on the mechanisms and stages of adipose tissue development and function. In this review, we focus on the molecular regulation of white adipose tissue development, from commitment to terminal differentiation, and examine key functional aspects of WAT that are crucial for normal physiology and systemic metabolic homeostasis.
Collapse
Affiliation(s)
- Presley D. Dowker-Key
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Praveen Kumar Jadi
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Nicholas B. Gill
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Katelin N. Hubbard
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Ahmed Elshaarrawi
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Naba D. Alfatlawy
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Ahmed Bettaieb
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
- Graduate School of Genome Science and Technology, University of Tennessee, Knoxville, TN 37996-0840, USA
- Department of Biochemistry, Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996-0840, USA
| |
Collapse
|
31
|
Frankovic I, Djuricic I, Ninic A, Vekic J, Vorkapic T, Erceg S, Gojkovic T, Tomasevic R, Mamic M, Mitrovic M, Zeljkovic A. Increased Odds of Metabolic Dysfunction-Associated Steatotic Liver Disease Are Linked to Reduced n-6, but Not n-3 Polyunsaturated Fatty Acids in Plasma. Biomolecules 2024; 14:902. [PMID: 39199290 PMCID: PMC11353166 DOI: 10.3390/biom14080902] [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: 06/25/2024] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 09/01/2024] Open
Abstract
The increasing prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) underscores the need for better understanding of its complex pathogenesis. Lipid accumulation in hepatocytes is among principal mechanisms contributing to MASLD development. While routine lipid parameters are well studied, the profile of circulating fatty acids in MASLD patients remains less explored. This study aimed to assess relative proportions of individual fatty acids in plasma of MASLD patients and to explore their associations with other biochemical markers of MASLD. Ninety-one patients and 48 healthy individuals were enrolled. The relative proportions of fatty acids in plasma were determined using gas chromatography with FID detection. Proportions of total n-6 polyunsaturated fatty acids (PUFAs) and linoleic acid (LA) in plasma were lower in MASLD patients (p = 0.001 and p = 0.004, respectively), with no differences observed in n-3 PUFAs. Total plasma n-6 PUFAs correlated negatively with body mass index, hepatic steatosis indices, triglyceride concentration and coronary risk index. Decreased prevalence of n-6 PUFAs in plasma was independently associated with higher odds of MASLD (OR = 0.769; CI: 0.611-0.968; p = 0.025). Our findings indicate an altered circulatory fatty acid distribution in MASLD, characterized by a reduced amount of n-6 PUFAs, particularly LA, which may have significant implications for the prevention and treatment of MASLD.
Collapse
Affiliation(s)
- Irena Frankovic
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia; (I.F.); (I.D.); (A.N.); (J.V.); (T.V.); (S.E.); (T.G.)
| | - Ivana Djuricic
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia; (I.F.); (I.D.); (A.N.); (J.V.); (T.V.); (S.E.); (T.G.)
| | - Ana Ninic
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia; (I.F.); (I.D.); (A.N.); (J.V.); (T.V.); (S.E.); (T.G.)
| | - Jelena Vekic
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia; (I.F.); (I.D.); (A.N.); (J.V.); (T.V.); (S.E.); (T.G.)
| | - Tara Vorkapic
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia; (I.F.); (I.D.); (A.N.); (J.V.); (T.V.); (S.E.); (T.G.)
| | - Sanja Erceg
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia; (I.F.); (I.D.); (A.N.); (J.V.); (T.V.); (S.E.); (T.G.)
| | - Tamara Gojkovic
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia; (I.F.); (I.D.); (A.N.); (J.V.); (T.V.); (S.E.); (T.G.)
| | - Ratko Tomasevic
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
- Department of Gastroenterology and Hepatology, Clinic for Internal Medicine, Clinical Hospital Center Zemun, 11080 Belgrade, Serbia
| | - Milica Mamic
- Department of Laboratory Diagnostics, Clinical Hospital Center Zemun, 11080 Belgrade, Serbia;
| | - Milos Mitrovic
- Clinical Department for Gastroenterology and Hepatology, University Medical Center Zvezdara, 11000 Belgrade, Serbia;
| | - Aleksandra Zeljkovic
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia; (I.F.); (I.D.); (A.N.); (J.V.); (T.V.); (S.E.); (T.G.)
| |
Collapse
|
32
|
Raymond J, Morin A, Bradley-Garcia M, Plamondon H. Juvenile/Peripubertal Exposure to Omega-3 and Environmental Enrichment Differentially Affects CORT Secretion and Adulthood Stress Coping, Sociability, and CA3 Glucocorticoid Receptor Expression in Male and Female Rats. Nutrients 2024; 16:2350. [PMID: 39064793 PMCID: PMC11279577 DOI: 10.3390/nu16142350] [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: 06/17/2024] [Revised: 07/11/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
In adult rats, omega-3 supplementation through fish oil (FO) and environmental enrichment (EE) have shown beneficial effects on cognition and stress regulation. This study assessed sex-specific effects of FO and EE during adolescence, a period critical for brain maturation, on adulthood coping mechanisms, sociability, and glucocorticoid regulation. An amount of 64 Wistar rats [n = 32/sex; postnatal day (PND) 23] were assigned to supplementation of control soybean oil (CSO) or menhaden fish oil (FO; 0.3 mL/100 g) from PND28 to 47 and exposed to EE or regular cage (RC) housing from PND28 to 58, with their blood corticosterone (CORT) levels being assessed weekly. As adults, exposure to repeated forced swim tests (FSTs; PND90-91) enabled analysis of coping responses, while socioemotional and memory responses were evaluated using the OFT, EPM, SIT, and Y maze tests (PND92-94). Immunohistochemistry determined hippocampal CA1/CA3 glucocorticoid receptor (GR) expression (PND95). CORT secretion gradually increased as the supplementation period elapsed in female rats, while changes were minimal in males. Coping strategies in the FST differed between sexes, particularly in FO-fed rats, where females and males, respectively, favoured floating and tail support to minimise energy consumption and maintain immobility. In the SIT, FO/EE promoted sociability in females, while a CSO diet favoured social recognition in males. Reduced CA3 GR-ir expression was found in FO/RC and CSO/EE rat groups, supporting stress resilience and memory consolidation. Our findings support environment and dietary conditions to exert a sex-specific impact on biobehavioural responses.
Collapse
Affiliation(s)
| | | | | | - Hélène Plamondon
- Behavioural Neuroscience Group, School of Psychology, University of Ottawa, 136 Jean-Jacques Lussier, Ottawa, ON K1N 6N5, Canada; (J.R.); (A.M.); (M.B.-G.)
| |
Collapse
|
33
|
Parchem K, Letsiou S, Petan T, Oskolkova O, Medina I, Kuda O, O'Donnell VB, Nicolaou A, Fedorova M, Bochkov V, Gladine C. Oxylipin profiling for clinical research: Current status and future perspectives. Prog Lipid Res 2024; 95:101276. [PMID: 38697517 DOI: 10.1016/j.plipres.2024.101276] [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: 12/12/2023] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
Oxylipins are potent lipid mediators with increasing interest in clinical research. They are usually measured in systemic circulation and can provide a wealth of information regarding key biological processes such as inflammation, vascular tone, or blood coagulation. Although procedures still require harmonization to generate comparable oxylipin datasets, performing comprehensive profiling of circulating oxylipins in large studies is feasible and no longer restricted by technical barriers. However, it is essential to improve and facilitate the biological interpretation of complex oxylipin profiles to truly leverage their potential in clinical research. This requires regular updating of our knowledge about the metabolism and the mode of action of oxylipins, and consideration of all factors that may influence circulating oxylipin profiles independently of the studied disease or condition. This review aims to provide the readers with updated and necessary information regarding oxylipin metabolism, their different forms in systemic circulation, the current limitations in deducing oxylipin cellular effects from in vitro bioactivity studies, the biological and technical confounding factors needed to consider for a proper interpretation of oxylipin profiles.
Collapse
Affiliation(s)
- Karol Parchem
- Department of Food Chemistry, Technology and Biotechnology, Faculty of Chemistry, Gdańsk University of Technology, 11/12 Gabriela Narutowicza St., 80-233 Gdańsk, Poland; Department of Analytical Chemistry, Faculty of Chemical Technology, University of Pardubice, Studentská 573, 53210 Pardubice, Czech Republic.
| | - Sophia Letsiou
- Department of Biomedical Sciences, University of West Attica, Ag. Spiridonos St. Egaleo, 12243 Athens, Greece.
| | - Toni Petan
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Jamova cesta 39, SI-1000 Ljubljana, Slovenia.
| | - Olga Oskolkova
- Institute of Pharmaceutical Sciences, University of Graz, Humboldtstrasse 46/III, 8010 Graz, Austria.
| | - Isabel Medina
- Instituto de Investigaciones Marinas-Consejo Superior de Investigaciones Científicas (IIM-CSIC), Eduardo Cabello 6, E-36208 Vigo, Spain.
| | - Ondrej Kuda
- Institute of Physiology, Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czech Republic.
| | - Valerie B O'Donnell
- Systems Immunity Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK.
| | - Anna Nicolaou
- School of Health Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9NT, UK.
| | - Maria Fedorova
- Center of Membrane Biochemistry and Lipid Research, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, 01307 Dresden, Germany.
| | - Valery Bochkov
- Institute of Pharmaceutical Sciences, University of Graz, Humboldtstrasse 46/III, 8010 Graz, Austria.
| | - Cécile Gladine
- Université Clermont Auvergne, INRAE, UNH, Clermont-Ferrand, France.
| |
Collapse
|
34
|
Żulińska S, Strosznajder AK, Strosznajder JB. Current View on PPAR-α and Its Relation to Neurosteroids in Alzheimer's Disease and Other Neuropsychiatric Disorders: Promising Targets in a Therapeutic Strategy. Int J Mol Sci 2024; 25:7106. [PMID: 39000217 PMCID: PMC11241121 DOI: 10.3390/ijms25137106] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/19/2024] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) may play an important role in the pathomechanism/pathogenesis of Alzheimer's disease (AD) and several other neurological/neuropsychiatric disorders. AD leads to progressive alterations in the redox state, ion homeostasis, lipids, and protein metabolism. Significant alterations in molecular processes and the functioning of several signaling pathways result in the degeneration and death of synapses and neuronal cells, leading to the most severe dementia. Peroxisome proliferator-activated receptor alpha (PPAR-α) is among the processes affected by AD; it regulates the transcription of genes related to the metabolism of cholesterol, fatty acids, other lipids and neurotransmission, mitochondria biogenesis, and function. PPAR-α is involved in the cholesterol transport to mitochondria, the substrate for neurosteroid biosynthesis. PPAR-α-coding enzymes, such as sulfotransferases, which are responsible for neurosteroid sulfation. The relation between PPAR-α and cholesterol/neurosteroids may have a significant impact on the course and progression of neurodegeneration/neuroprotection processes. Unfortunately, despite many years of intensive studies, the pathogenesis of AD is unknown and therapy for AD and other neurodegenerative diseases is symptomatic, presenting a significant goal and challenge today. This review presents recent achievements in therapeutic approaches for AD, which are targeting PPAR-α and its relation to cholesterol and neurosteroids in AD and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Sylwia Żulińska
- Department of Cellular Signaling, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego St., 02-106 Warsaw, Poland;
| | - Anna K. Strosznajder
- Department of Psychiatry, Medical University of Warsaw, Nowowiejska St. 27, 00-665 Warsaw, Poland;
| | - Joanna B. Strosznajder
- Department of Cellular Signaling, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego St., 02-106 Warsaw, Poland;
| |
Collapse
|
35
|
Zhao Y, Zhao X, Feng X. Alpha-lipoic acid upregulates the PPARγ/NRF2/GPX4 signal pathway to inhibit ferroptosis in the pathogenesis of unexplained recurrent pregnancy loss. Open Med (Wars) 2024; 19:20240963. [PMID: 38859880 PMCID: PMC11163161 DOI: 10.1515/med-2024-0963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/16/2024] [Accepted: 04/07/2024] [Indexed: 06/12/2024] Open
Abstract
Aim With unknown etiology and limited treatment options, unexplained recurrent pregnancy loss (URPL) remains a thorny problem. Ferroptosis, a newly identified type of cell death, has been shown to be crucial in the development in reproductive disorders. This study aims to explore the specific mechanism of ferroptosis in URPL and to uncover whether alpha-lipoic acid (ALA) can inhibit ferroptosis, and then exert a protective effect in URPL. Method The decidua tissues of URPL and control patients who actively terminated pregnancy were collected. The CBA/J × DBA/2 murine models of URPL were established, and were randomly treated with peroxisome proliferator activated receptor γ (PPARγ) agonists (Rosiglitazone) and ALA. The CBA/J × BALB/c murine models of normal pregnancy were intraperitoneally injected with PPARγ inhibitors (T0070907). Here, we used reactive oxygen species (ROS), malondialdehyde (MDA), glutathione (GSH)/GSSG, and FeRhoNox-1 analysis to detect the level of ferroptosis. We used quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR) analysis to evaluate the mRNA level of PPARγ. Besides, western blot and immunofluorescence were utilized to test the expression profile of PPARγ/nuclear factor erythroid 2-related factor 2 (NRF2)/glutathione peroxidase 4 (GPX4). Results In this study, we found that iron deposition was increased in the decidual tissue of patients with URPL. Additionally, the changes in cell morphology, the level of ROS, MDA, GSH, and the expression of ferroptosis marker proteins NRF2/GPX4 confirmed activated ferroptosis in URPL. Besides, bioinformatics analysis combined with experiments confirmed that PPARγ was critical in triggering NRF2/GPX4 pathway in URPL. Furthermore, URPL mouse models were established, and the results showed that PPARγ/NRF2/GPX4-mediated ferroptosis was also significantly increased, which could be mitigated by ALA treatment. Conclusion Overall, these findings suggest that ferroptosis may play an important role in URPL, and ALA might be a promising therapeutic drug for improving pregnancy outcomes in URPL via targeting the PPARγ/NRF2/GPX4 pathway.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Gynecology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Xiaoxuan Zhao
- Department of Traditional Chinese Medicine (TCM) Gynecology, Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310007, China
| | - Xiaoling Feng
- Department of Gynecology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| |
Collapse
|
36
|
Zhang YA, Li FW, Dong YX, Xie WJ, Wang HB. PPAR-γ regulates the polarization of M2 macrophages to improve the microenvironment for autologous fat grafting. FASEB J 2024; 38:e23613. [PMID: 38661048 DOI: 10.1096/fj.202400126r] [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: 01/17/2024] [Revised: 03/19/2024] [Accepted: 04/02/2024] [Indexed: 04/26/2024]
Abstract
The unpredictable survival rate of autologous fat grafting (AFG) seriously affects its clinical application. Improving the survival rate of AFG has become an unresolved issue in plastic surgery. Peroxisome proliferator-activated receptor-γ (PPAR-γ) regulates the adipogenic differentiation of adipocytes, but the functional mechanism in AFG remains unclear. In this study, we established an animal model of AFG and demonstrated the superior therapeutic effect of PPAR-γ regulation in the process of AFG. From day 3 after fat grafting, the PPAR-γ agonist rosiglitazone group consistently showed better adipose integrity, fewer oil cysts, and fibrosis. Massive macrophage infiltration was observed after 7 days. At the same time, M2 macrophages begin to appear. At day 14, M2 macrophages gradually became the dominant cell population, which suppressed inflammation and promoted revascularization and fat regeneration. In addition, transcriptome sequencing showed that the differentially expressed genes in the Rosiglitazone group were associated with the pathways of adipose regeneration, differentiation, and angiogenesis; these results provide new ideas for clinical treatment.
Collapse
Affiliation(s)
- Ya-An Zhang
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Fang-Wei Li
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Yun-Xian Dong
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Wen-Jie Xie
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Hai-Bin Wang
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, Guangzhou, China
| |
Collapse
|
37
|
Amoah AS, Pestov NB, Korneenko TV, Prokhorenko IA, Kurakin GF, Barlev NA. Lipoxygenases at the Intersection of Infection and Carcinogenesis. Int J Mol Sci 2024; 25:3961. [PMID: 38612771 PMCID: PMC11011848 DOI: 10.3390/ijms25073961] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/08/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
The persisting presence of opportunistic pathogens like Pseudomonas aeruginosa poses a significant threat to many immunocompromised cancer patients with pulmonary infections. This review highlights the complexity of interactions in the host's defensive eicosanoid signaling network and its hijacking by pathogenic bacteria to their own advantage. Human lipoxygenases (ALOXs) and their mouse counterparts are integral elements of the innate immune system, mostly operating in the pro-inflammatory mode. Taking into account the indispensable role of inflammation in carcinogenesis, lipoxygenases have counteracting roles in this process. In addition to describing the structure-function of lipoxygenases in this review, we discuss their roles in such critical processes as cancer cell signaling, metastases, death of cancer and immune cells through ferroptosis, as well as the roles of ALOXs in carcinogenesis promoted by pathogenic infections. Finally, we discuss perspectives of novel oncotherapeutic approaches to harness lipoxygenase signaling in tumors.
Collapse
Affiliation(s)
- Abdul-Saleem Amoah
- Institute of Biomedical Chemistry, Moscow 119121, Russia; (A.-S.A.); (N.A.B.)
- Laboratory of Molecular Oncology, Phystech School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
| | - Nikolay B. Pestov
- Institute of Biomedical Chemistry, Moscow 119121, Russia; (A.-S.A.); (N.A.B.)
- Group of Cross-Linking Enzymes, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (T.V.K.); (I.A.P.)
- Laboratory of Tick-Borne Encephalitis and Other Viral Encephalitides, Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products, Moscow 108819, Russia
- Vavilov Institute of General Genetics, Moscow 119991, Russia
| | - Tatyana V. Korneenko
- Group of Cross-Linking Enzymes, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (T.V.K.); (I.A.P.)
| | - Igor A. Prokhorenko
- Group of Cross-Linking Enzymes, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (T.V.K.); (I.A.P.)
| | - Georgy F. Kurakin
- Department of Biochemistry, Pirogov Russian National Research Medical University, Moscow 117513, Russia;
| | - Nickolai A. Barlev
- Institute of Biomedical Chemistry, Moscow 119121, Russia; (A.-S.A.); (N.A.B.)
- Laboratory of Tick-Borne Encephalitis and Other Viral Encephalitides, Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products, Moscow 108819, Russia
| |
Collapse
|
38
|
Bu Y, Yang S, Wang D, Hu S, Zhang Q, Wu Z, Yang C. Role of soluble epoxide hydrolase in pain and depression comorbidity. Neurobiol Dis 2024; 193:106443. [PMID: 38395315 DOI: 10.1016/j.nbd.2024.106443] [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: 09/18/2023] [Revised: 02/15/2024] [Accepted: 02/19/2024] [Indexed: 02/25/2024] Open
Abstract
The coexistence of chronic pain and depression in clinical practice places a substantial social burden and profoundly impacts in patients. Although a clear correlation exists, the underlying mechanism of comorbidity between chronic pain and depression remains elusive. Research conducted in recent decades has uncovered that soluble epoxide hydrolase, a pivotal enzyme in the metabolism of polyunsaturated fatty acids, plays a crucial role in inflammation. Interestingly, this enzyme is intricately linked to the development of both pain and depression. With this understanding, this review aims to summarize the roles of soluble epoxide hydrolase in pain, depression, and their comorbidity. Simultaneously, we will also explore the underlying mechanisms, providing guidance for future research and drug development.
Collapse
Affiliation(s)
- Yuchen Bu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Siqi Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Di Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Suwan Hu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Qi Zhang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zifeng Wu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Chun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
39
|
Trimarco A, Audano M, Marca RL, Cariello M, Falco M, Pedretti S, Imperato G, Cestaro A, Podini P, Dina G, Quattrini A, Massimino L, Caruso D, Mitro N, Taveggia C. Prostaglandin D2 synthase controls Schwann cells metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.29.582775. [PMID: 38496560 PMCID: PMC10942270 DOI: 10.1101/2024.02.29.582775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
We previously reported that in the absence of Prostaglandin D2 synthase (L-PGDS) peripheral nerves are hypomyelinated in development and that with aging they present aberrant myelin sheaths. We now demonstrate that L-PGDS expressed in Schwann cells is part of a coordinated program aiming at preserving myelin integrity. In vivo and in vitro lipidomic, metabolomic and transcriptomic analyses confirmed that myelin lipids composition, Schwann cells energetic metabolism and key enzymes controlling these processes are altered in the absence of L-PGDS. Moreover, Schwann cells undergo a metabolic rewiring and turn to acetate as the main energetic source. Further, they produce ketone bodies to ensure glial cell and neuronal survival. Importantly, we demonstrate that all these changes correlate with morphological myelin alterations and describe the first physiological pathway implicated in preserving PNS myelin. Collectively, we posit that myelin lipids serve as a reservoir to provide ketone bodies, which together with acetate represent the adaptive substrates Schwann cells can rely on to sustain the axo-glial unit and preserve the integrity of the PNS.
Collapse
|
40
|
Carter WA, DeMoranville KJ, Trost L, Bryła A, Działo M, Sadowska ET, Bauchinger U, Pierce B, McWilliams SR. Dietary fatty acids and flight-training influence the expression of the eicosanoid hormone prostacyclin in songbirds. Comp Biochem Physiol A Mol Integr Physiol 2024; 288:111561. [PMID: 38056555 DOI: 10.1016/j.cbpa.2023.111561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 12/01/2023] [Accepted: 12/01/2023] [Indexed: 12/08/2023]
Abstract
Diet shifts can alter tissue fatty acid composition in birds, which is subsequently related to metabolic patterns. Eicosanoids, short-lived fatty acid-derived hormones, have been proposed to mediate these relationships but neither baseline concentrations nor the responses to diet and exercise have been measured in songbirds. We quantified a stable derivative of the vasodilatory eicosanoid prostacyclin in the plasma of male European Starlings (Sturnus vulgaris, N = 25) fed semisynthetic diets with either high (PUFA) or low (MUFA) amounts of n6 fatty acid precursors to prostacyclin. Plasma samples were taken from each bird before, immediately after, and two days following a 15-day flight-training regimen that a subset of birds (N = 17) underwent. We found elevated prostacyclin levels in flight-trained birds fed the PUFA diet compared to those fed the MUFA diet and a positive relationship between prostacyclin and body condition, indexed by fat score. Prostacyclin concentrations also significantly decreased at the final time point. These results are consistent with the proposed influences of precursor availability (i.e., dietary fatty acids) and regulatory feedback associated with exercise (i.e., fuel supply and inflammation), and suggest that prostacyclin may be an important mediator of dietary influence on songbird physiology.
Collapse
Affiliation(s)
- Wales A Carter
- Department of Resources Science, University of Rhode Island, Kingston, RI, USA.
| | | | - Lisa Trost
- Department for Behavioural Neurobiology, Max Planck Institute for Ornithology, Seewiesen, Germany
| | - Amadeusz Bryła
- Institute of Environmental Sciences, Jagiellonian University, Kraków, Poland
| | - Maciej Działo
- Institute of Environmental Sciences, Jagiellonian University, Kraków, Poland
| | - Edyta T Sadowska
- Institute of Environmental Sciences, Jagiellonian University, Kraków, Poland
| | - Ulf Bauchinger
- Institute of Environmental Sciences, Jagiellonian University, Kraków, Poland
| | - Barbara Pierce
- Department of Biology, Sacred Heart University, Fairfield, CT, USA
| | - Scott R McWilliams
- Department of Resources Science, University of Rhode Island, Kingston, RI, USA
| |
Collapse
|
41
|
Fan J, Liu C, Zhao Y, Xu Q, Yin Z, Liu Z, Mu Y. Single-Cell RNA Sequencing Reveals Differences in Chromatin Remodeling and Energy Metabolism among In Vivo-Developed, In Vitro-Fertilized, and Parthenogenetically Activated Embryos from the Oocyte to 8-Cell Stages in Pigs. Animals (Basel) 2024; 14:465. [PMID: 38338108 PMCID: PMC10854501 DOI: 10.3390/ani14030465] [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: 12/19/2023] [Revised: 01/26/2024] [Accepted: 01/28/2024] [Indexed: 02/12/2024] Open
Abstract
In vitro-fertilized (IVF) and parthenogenetically activated (PA) embryos, key to genetic engineering, face more developmental challenges than in vivo-developed embryos (IVV). We analyzed single-cell RNA-seq data from the oocyte to eight-cell stages in IVV, IVF, and PA porcine embryos, focusing on developmental differences during early zygotic genome activation (ZGA), a vital stage for embryonic development. (1) Our findings reveal that in vitro embryos (IVF and PA) exhibit more similar developmental trajectories compared to IVV embryos, with PA embryos showing the least gene diversity at each stage. (2) Significant differences in maternal mRNA, particularly affecting mRNA splicing, energy metabolism, and chromatin remodeling, were observed. Key genes like SMARCB1 (in vivo) and SIRT1 (in vitro) played major roles, with HDAC1 (in vivo) and EZH2 (in vitro) likely central in their complexes. (3) Across different types of embryos, there was minimal overlap in gene upregulation during ZGA, with IVV embryos demonstrating more pronounced upregulation. During minor ZGA, global epigenetic modification patterns diverged and expanded further. Specifically, in IVV, genes, especially those linked to H4 acetylation and H2 ubiquitination, were more actively regulated compared to PA embryos, which showed an increase in H3 methylation. Additionally, both types displayed a distinction in DNA methylation. During major ZGA, IVV distinctively upregulated genes related to mitochondrial regulation, ATP synthesis, and oxidative phosphorylation. (4) Furthermore, disparities in mRNA degradation-related genes between in vivo and in vitro embryos were more pronounced during major ZGA. In IVV, there was significant maternal mRNA degradation. Maternal genes regulating phosphatase activity and cell junctions, highly expressed in both in vivo and in vitro embryos, were degraded in IVV in a timely manner but not in in vitro embryos. (5) Our analysis also highlighted a higher expression of many mitochondrially encoded genes in in vitro embryos, yet their nucleosome occupancy and the ATP8 expression were notably higher in IVV.
Collapse
Affiliation(s)
- Jianlin Fan
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (J.F.); (C.L.); (Y.Z.); (Q.X.); (Z.Y.)
- College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Chang Liu
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (J.F.); (C.L.); (Y.Z.); (Q.X.); (Z.Y.)
- College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Yunjing Zhao
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (J.F.); (C.L.); (Y.Z.); (Q.X.); (Z.Y.)
| | - Qianqian Xu
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (J.F.); (C.L.); (Y.Z.); (Q.X.); (Z.Y.)
- College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Zhi Yin
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (J.F.); (C.L.); (Y.Z.); (Q.X.); (Z.Y.)
- College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Zhonghua Liu
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (J.F.); (C.L.); (Y.Z.); (Q.X.); (Z.Y.)
- College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Yanshuang Mu
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (J.F.); (C.L.); (Y.Z.); (Q.X.); (Z.Y.)
- College of Life Science, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
42
|
Ardenkjær-Skinnerup J, Nissen ACVE, Nikolov NG, Hadrup N, Ravn-Haren G, Wedebye EB, Vogel U. Orthogonal assay and QSAR modelling of Tox21 PPARγ antagonist in vitro high-throughput screening assay. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 105:104347. [PMID: 38143042 DOI: 10.1016/j.etap.2023.104347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/18/2023] [Indexed: 12/26/2023]
Abstract
Disruption of signalling mediated by the nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ) is associated with risk of cancer, metabolic diseases, and endocrine disruption. The purpose of this study was to identify environmental chemicals acting as PPARγ antagonists. Data from the Tox21 PPARγ antagonism assay were replicated using a reporter system in HEK293 cells. Two quantitative structure-activity relationship (QSAR) models were developed, and five REACH-registered substances predicted positive were tested in vitro. Reporter assay results were consistent with Tox21 data since all conflicting results could be explained by assay interference. QSAR models showed good predictive performance, and follow-up experiments revealed two PPARγ antagonists out of three non-interfering chemicals. In conclusion, the developed QSAR models and follow-up experiments are important steps in the discovery of potential endocrine- and metabolism-disrupting chemicals.
Collapse
Affiliation(s)
- Jacob Ardenkjær-Skinnerup
- The National Food Institute, Technical University of Denmark, Kemitorvet 202, 2800 Kongens Lyngby, Denmark; The National Research Centre for the Working Environment, Lersø Parkallé 105, 2100 Copenhagen Ø, Denmark
| | | | - Nikolai Georgiev Nikolov
- The National Food Institute, Technical University of Denmark, Kemitorvet 202, 2800 Kongens Lyngby, Denmark
| | - Niels Hadrup
- The National Food Institute, Technical University of Denmark, Kemitorvet 202, 2800 Kongens Lyngby, Denmark; The National Research Centre for the Working Environment, Lersø Parkallé 105, 2100 Copenhagen Ø, Denmark
| | - Gitte Ravn-Haren
- The National Food Institute, Technical University of Denmark, Kemitorvet 202, 2800 Kongens Lyngby, Denmark
| | - Eva Bay Wedebye
- The National Food Institute, Technical University of Denmark, Kemitorvet 202, 2800 Kongens Lyngby, Denmark
| | - Ulla Vogel
- The National Food Institute, Technical University of Denmark, Kemitorvet 202, 2800 Kongens Lyngby, Denmark; The National Research Centre for the Working Environment, Lersø Parkallé 105, 2100 Copenhagen Ø, Denmark.
| |
Collapse
|
43
|
Ma XZ, Chen LL, Qu L, Li H, Wang J, Song N, Xie JX. Gut microbiota-induced CXCL1 elevation triggers early neuroinflammation in the substantia nigra of Parkinsonian mice. Acta Pharmacol Sin 2024; 45:52-65. [PMID: 37674043 PMCID: PMC10770039 DOI: 10.1038/s41401-023-01147-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 07/25/2023] [Indexed: 09/08/2023]
Abstract
Gut microbiota disturbance and systemic inflammation have been implicated in the degeneration of dopaminergic neurons in Parkinson's disease (PD). How the alteration of gut microbiota results in neuropathological events in PD remains elusive. In this study, we explored whether and how environmental insults caused early neuropathological events in the substantia nigra (SN) of a PD mouse model. Aged (12-month-old) mice were orally administered rotenone (6.25 mg·kg-1·d-1) 5 days per week for 2 months. We demonstrated that oral administration of rotenone to ageing mice was sufficient to establish a PD mouse model and that microglial activation and iron deposition selectively appeared in the SN of the mice prior to loss of motor coordination and dopaminergic neurons, and these events could be fully blocked by microglial elimination with a PLX5622-formulated diet. 16 S rDNA sequencing analysis showed that the gut microbiota in rotenone-treated mice was altered, and mice receiving faecal microbial transplantation (FMT) from ageing mice treated with rotenone for 2 months exhibited the same pathology in the SN. We demonstrated that C-X-C motif chemokine ligand-1 (CXCL1) was an essential molecule, as intravenous injection of CXCL1 mimicked almost all the pathology in serum and SN induced by oral rotenone and FMT. Using metabolomics and transcriptomics analyses, we identified the PPAR pathway as a key pathway involved in rotenone-induced neuronal damage. Inhibition of the PPARγ pathway was consistent in the above models, whereas its activation by linoleic acid (60 mg·kg-1·d-1, i.g. for 1 week) could block these pathological events in mice intravenously injected with CXCL1. Altogether, these results reveal that the altered gut microbiota resulted in neuroinflammation and iron deposition occurring early in the SN of ageing mice with oral administration of rotenone, much earlier than motor symptoms and dopaminergic neuron loss. We found that CXCL1 plays a crucial role in this process, possibly via PPARγ signalling inhibition. This study may pave the way for understanding the "brain-gut-microbiota" molecular regulatory networks in PD pathogenesis. The aged C57BL/6 male mice with rotenone intragastric administration showed altered gut microbiota, which caused systemic inflammation, PPARγ signalling inhibition and neuroinflammation, brain iron deposition and ferroptosis, and eventually dopaminergic neurodegeneration in PD.
Collapse
Affiliation(s)
- Xi-Zhen Ma
- Institute of Brain Science and Disease, School of Basic Medicine, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266021, China
| | - Lei-Lei Chen
- Institute of Brain Science and Disease, School of Basic Medicine, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266021, China
| | - Le Qu
- Institute of Brain Science and Disease, School of Basic Medicine, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266021, China
| | - Hui Li
- Institute of Brain Science and Disease, School of Basic Medicine, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266021, China
| | - Jun Wang
- Institute of Brain Science and Disease, School of Basic Medicine, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266021, China
| | - Ning Song
- Institute of Brain Science and Disease, School of Basic Medicine, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266021, China.
| | - Jun-Xia Xie
- Institute of Brain Science and Disease, School of Basic Medicine, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266021, China.
| |
Collapse
|
44
|
Sheremeta CL, Yarlagadda S, Smythe ML, Noakes PG. Prostaglandins in the Inflamed Central Nervous System: Potential Therapeutic Targets. Curr Drug Targets 2024; 25:885-908. [PMID: 39177131 PMCID: PMC11774313 DOI: 10.2174/0113894501323980240815113851] [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: 04/29/2024] [Revised: 07/11/2024] [Accepted: 07/19/2024] [Indexed: 08/24/2024]
Abstract
The global burden of neurological disorders is evident, yet there remains limited efficacious therapeutics for their treatment. There is a growing recognition of the role of inflammation in diseases of the central nervous system (CNS); among the numerous inflammatory mediators involved, prostaglandins play a crucial role. Prostaglandins are small lipid mediators derived from arachidonic acid via multi-enzymatic pathways. The actions of prostaglandins are varied, with each prostaglandin having a specific role in maintaining homeostasis. In the CNS, prostaglandins can have neuroprotective or neurotoxic properties depending on their specific G-protein receptor. These G-protein receptors have varying subfamilies, tissue distribution, and signal transduction cascades. Further studies into the impact of prostaglandins in CNS-based diseases may contribute to the clarification of their actions, hopefully leading to the development of efficacious therapeutic strategies. This review focuses on the roles played by prostaglandins in neural degeneration, with a focus on Alzheimer's Disease, Multiple Sclerosis, and Amyotrophic Lateral Sclerosis in both preclinical and clinical settings. We further discuss current prostaglandin-related agonists and antagonists concerning suggestions for their use as future therapeutics.
Collapse
Affiliation(s)
- Chynna-Loren Sheremeta
- Institute for Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia
- School of Biomedical Sciences, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Sai Yarlagadda
- Institute for Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia
- School of Biomedical Sciences, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Mark L. Smythe
- Institute for Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Peter G. Noakes
- School of Biomedical Sciences, The University of Queensland, St. Lucia, QLD 4072, Australia
- Queensland Brain Institute, The University of Queensland, St. Lucia, QLD 4072, Australia
| |
Collapse
|
45
|
Qiu Y, Gan M, Wang X, Liao T, Chen Q, Lei Y, Chen L, Wang J, Zhao Y, Niu L, Wang Y, Zhang S, Zhu L, Shen L. The global perspective on peroxisome proliferator-activated receptor γ (PPARγ) in ectopic fat deposition: A review. Int J Biol Macromol 2023; 253:127042. [PMID: 37742894 DOI: 10.1016/j.ijbiomac.2023.127042] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 09/26/2023]
Abstract
Excessive expansion of adipocytes can have unhealthy consequences as excess free fatty acids enter other tissues and cause ectopic fat deposition by resynthesizing triglycerides. This lipid accumulation in various tissues is harmful and can increase the risk of related metabolic diseases such as type II diabetes, cardiovascular disease, and insulin resistance. Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear hormone receptor superfamily that play a key role in energy metabolism as fatty acid metabolism sensors, and peroxisome proliferator-activated receptor γ (PPARγ) is the main subtype responsible for fat cell differentiation and adipogenesis. In this paper, we introduce the main structure and function of PPARγ and its regulatory role in the process of lipogenesis in the liver, kidney, skeletal muscle, and pancreas. This information can serve as a reference for further understanding the regulatory mechanisms and measures of the PPAR family in the process of ectopic fat deposition.
Collapse
Affiliation(s)
- Yanhao Qiu
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Mailin Gan
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Xingyu Wang
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Tianci Liao
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Qiuyang Chen
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Yuhang Lei
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Lei Chen
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Jinyong Wang
- Chongqing Academy of Animal Science, Rongchang, Chongqing 402460, China
| | - Ye Zhao
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Lili Niu
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Yan Wang
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Shunhua Zhang
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Li Zhu
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China.
| | - Linyuan Shen
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
46
|
Zhao R, Wu R, Jin J, Ning K, Wang Z, Yi X, Kapilevich L, Liu J. Signaling pathways regulated by natural active ingredients in the fight against exercise fatigue-a review. Front Pharmacol 2023; 14:1269878. [PMID: 38155906 PMCID: PMC10752993 DOI: 10.3389/fphar.2023.1269878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 12/04/2023] [Indexed: 12/30/2023] Open
Abstract
Exercise fatigue is a normal protective mechanism of the body. However, long-term fatigue hinders normal metabolism and exercise capacity. The generation and recovery from exercise fatigue involves alterations in multiple signaling pathways, mainly AMPK, PI3K/Akt, Nrf2/ARE, NF-κB, PINK1/Parkin, and BDNF/TrkB, as well as MAPK signaling pathways that mediate energy supply, reduction of metabolites, oxidative stress homeostasis, muscle fiber type switching, and central protective effects. In recent studies, a rich variety of natural active ingredients have been identified in traditional Chinese medicines and plant extracts with anti-fatigue effects, opening up the field of research in new anti-fatigue drugs. In this review we give an overview of the signaling pathways associated with the activity of natural food active ingredients against exercise fatigue. Such a comprehensive review is necessary to understand the potential of these materials as preventive measures and treatments of exercise fatigue. We expect the findings highlighted and discussed here will help guide the development of new health products and provide a theoretical and scientific basis for future research on exercise fatigue.
Collapse
Affiliation(s)
- Rongyue Zhao
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Ruomeng Wu
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Junjie Jin
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Ke Ning
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Zhuo Wang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Xuejie Yi
- Exercise and Health Research Center, Department of Kinesiology, Shenyang Sport University, Shenyang, Liaoning, China
| | - Leonid Kapilevich
- Faculty of Physical Education, Nаtionаl Reseаrch Tomsk Stаte University, Tomsk, Russia
| | - Jiao Liu
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| |
Collapse
|
47
|
Santamarina AB, Mennitti LV, de Souza EA, Mesquita LMDS, Noronha IH, Vasconcelos JRC, Prado CM, Pisani LP. A low-carbohydrate diet with different fatty acids' sources in the treatment of obesity: Impact on insulin resistance and adipogenesis. Clin Nutr 2023; 42:2381-2394. [PMID: 37862824 DOI: 10.1016/j.clnu.2023.09.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/01/2023] [Accepted: 09/23/2023] [Indexed: 10/22/2023]
Abstract
BACKGROUND The search for nutritional intervention strategies against obesity has grown, highlighting the low-carbohydrate diet model. However, little is known about the impact of the quality of fatty acids consumed in this diet. Thus, we aim to investigate the influence of fatty acid quality on dietary strategy on obesity. METHODS Male Swiss mice were diet-induced to obesity. Afterward, mice consume a low-carb diet with different types of fat: saturated, polyunsaturated ω-3, ω-6, and monounsaturated ω-9 fatty acids. Weight gain and food consumption were monitored weekly. An oral glucose tolerance test was performed and blood and tissue samples were collected for analysis of insulin resistance markers. Protein expression of insulin signaling pathway molecules, lipid metabolism, mitochondrial function, macrophage polarization, and cytokine production were analyzed. RESULTS The high-fat diet was able to induce obesity and glucose intolerance. The switch to a low-carbohydrate dietary pattern reversed the glucose intolerance, with better results in the ω-3 and ω-9 groups. After the low-carbohydrate diet, groups ω-3 and ω-9 presented improved fasting serum glucose, insulin, and HOMA indexes. The low-carbohydrate diet also increased the activity of insulin pathway proteins such as IR, IRS1, and AKT. Furthermore, the ω-3 diet group showed greater activity of mitochondrial complexes and AMPK signaling pathway proteins. The ω-6 and ω-9 -rich diet induced M2-type macrophage polarization, as well as cytokine production modulation by the low-carbohydrate diet in the ω-3 and ω-9 groups. CONCLUSIONS Consuming a low-carbohydrate diet pattern promotes weight loss and improves glucose intolerance in obesity. Also, the quality of lipids has a direct influence, demonstrating that the consumption of ω-3 polyunsaturated and ω-9 monounsaturated lipids can lead to more favorable outcomes for the improvement of glucose intolerance, lipid metabolism, and anti-inflammatory effects.
Collapse
Affiliation(s)
- Aline B Santamarina
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo, Campus Baixada Santista - UNIFESP, Santos, São Paulo, Brazil
| | - Laís V Mennitti
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo, Campus Baixada Santista - UNIFESP, Santos, São Paulo, Brazil
| | - Esther A de Souza
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo, Campus Baixada Santista - UNIFESP, Santos, São Paulo, Brazil
| | - Leonardo M de Souza Mesquita
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas, Rua Pedro Zaccaria 1300, 13484-350 Limeira, São Paulo, Brazil
| | - Isaú H Noronha
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo, Campus Baixada Santista - UNIFESP, Santos, São Paulo, Brazil
| | - José Ronnie C Vasconcelos
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo, Campus Baixada Santista - UNIFESP, Santos, São Paulo, Brazil
| | - Carla M Prado
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo, Campus Baixada Santista - UNIFESP, Santos, São Paulo, Brazil
| | - Luciana P Pisani
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo, Campus Baixada Santista - UNIFESP, Santos, São Paulo, Brazil.
| |
Collapse
|
48
|
Uner B, Macit Celebi MS. Anti-obesity effects of chlorogenic acid and caffeine- lipid nanoparticles through PPAR-γ/C/EBP-ɑ pathways. Int J Obes (Lond) 2023; 47:1108-1119. [PMID: 37596386 DOI: 10.1038/s41366-023-01365-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/21/2023] [Accepted: 08/09/2023] [Indexed: 08/20/2023]
Abstract
Obesity is considered one of the most crucial health problems of the century. Therefore, reducing obesity is critically important. Caffeine (CF) and chlorogenic acid (CLA), which are substantial components in green bean coffee which maximize thermogenesis in brown adipose tissue. In our study, we have prepared CF, CLA, and CF + CLA loaded-solid lipid nanoparticles (SLN) since the SLNs are cost-effective, tissue-localized, and highly stable. The central composite design model was preferred to select the optimized formulation. UHPLC was used for quantification related to the CF and CLA amounts. The high-pressure homogenization (HPH) method was used while SLN formulations were prepared in the presence of poloxamer® 407 (surfactant) and Compritol® 888 ATO (solid lipid). The nanoparticles were characterized, followed by the utilization of 3T3-F442A cell lines for the evaluation of the adipogenesis activity of the formulations. Then, rt-PCR and ELISA studies of adipogenic markers were conducted. After optimal formulations were selected with an average of 110.2 ± 0.1 nm, CF (1 mM) + CLA (0.5 mM)-loaded SLN formulation has been proven significantly effective by using PPAR-γ/C/EBP-a pathways. In a nutshell, our study has shown that CF + CLA loaded-SLN has been affected 45.8% times more than regular extracted coffee (p < 0.05) on the adipocyte cells.
Collapse
Affiliation(s)
- Burcu Uner
- Department of Pharmaceutical and Administrative Sciences, University of Health Science and Pharmacy in St Louis, St. Louis, MO, USA.
| | | |
Collapse
|
49
|
Uner B, Macit Celebi MS. Anti-obesity effects of chlorogenic acid and caffeine- lipid nanoparticles through PPAR-γ/C/EBP-ɑ pathways. Int J Obes (Lond) 2023; 47:1108-1119. [DOI: 16.https:/doi.org/10.1038/s41366-023-01365-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/21/2023] [Accepted: 08/09/2023] [Indexed: 03/30/2025]
|
50
|
Yao CB, Feng L, Wu P, Liu Y, Jiang J, Zhang L, Mi HF, Zhou XQ, Jiang WD. Promotion of fatty acid metabolism and glucose metabolism in the muscle of sub-adult grass carp ( Ctenopharyngodon idella): The role of alpha-linoleic acid/linoleic acid (ALA/LNA) ratios. Food Chem X 2023; 19:100752. [PMID: 37384144 PMCID: PMC10293787 DOI: 10.1016/j.fochx.2023.100752] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 05/31/2023] [Accepted: 06/09/2023] [Indexed: 06/30/2023] Open
Abstract
The n6/n3 ratios improved meat quality of terrestrial animals, but alpha-linolenic acid/linoleic acid (ALA/LNA) ratios were rarely studied in aquatic animals. In this study, sub-adult grass carp (Ctenopharyngodon idella) were fed diets fed diets containing six varying ALA/LNA ratios (0.03, 0.47, 0.92, 1.33, 1.69, and 2.15) for 9 weeks and the total value of n3 + n6 (1.98) was kept constant for all six treatments. The results indicated optimal ALA/LNA ratio improved growth performance, changed fatty acid composition in grass carp muscle, and promoted glucose metabolism. Additionally, optimal ALA/LNA ratio improved chemical attributes by increasing crude protein and lipid contents, and technological attributes by increasing pH24h value and shear force in grass carp muscle. The signaling pathways related to fatty acid metabolism and glucose metabolism (LXRα/SREBP-1, PPARα, PPARγ, AMPK) might be responsible for these changes. Dietary optimal ALA/LNA ratio based on PWG, UFA and glucose contents was 1.03, 0.88 and 0.92, respectively.
Collapse
Affiliation(s)
- Chi-Bei Yao
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan 611130, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan 611130, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan 611130, China
| | - Jun Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Lu Zhang
- Tongwei Co., Ltd., Chengdu, China
- Healthy Aquaculture Key Laboratory of Sichuan Province, Sichuan 610041, China
| | | | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan 611130, China
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan 611130, China
| |
Collapse
|