1
|
Zhang LJ, Li SY, Fan SL, Nuerbiye A, Zhao JY, Keremu B, Yang L, Zhang K, Wang HY, Wang JH. Hepatoprotective activity of mulberry extract in NAFLD mice for regulating lipid metabolism and inflammation identified via AMPK/PPAR-γ/NF-κB axis. JOURNAL OF ETHNOPHARMACOLOGY 2025:120108. [PMID: 40490232 DOI: 10.1016/j.jep.2025.120108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 06/03/2025] [Accepted: 06/05/2025] [Indexed: 06/11/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Non-alcoholic fatty liver disease (NAFLD) is one of the most prevalent liver diseases worldwide, with an estimated global prevalence of 30%. An imbalance in lipid metabolism leads to the accumulation of lipotoxic lipids, inducing cellular stress, activating the NLRP3 inflammasome, and triggering apoptotic cell death. This cascade stimulates inflammation, driving NAFLD progression. Morus nigra L. is the only black mulberry species native to China. In traditional Uygur medicine, its fruit is valued for its hepatoprotective and lipid-lowering effects. AIM OF THE STUDY The mechanism by which mulberry extract (ME) alleviates NAFLD remains unclear. This study aimed to investigate the hepatoprotective and anti-inflammatory effects of ME. MATERIALS AND METHODS Network pharmacology was employed to predict the active components of ME and their potential target genes. Liver function markers (ALT, AST) and lipid profiles (TG, TC) were assessed using commercial assay kits. The therapeutic mechanism of ME against NAFLD was elucidated through an integrated approach combining transcriptomic and metabolomic analyses in a mouse NAFLD model. RESULTS UPLC-QTOF-MS analysis identified 131 active ingredients in ME. Network pharmacological analysis identified Akt1, Pparg, and Pparα as core targets. High-dose ME treatment markedly improved liver function, reducing ALT levels by 50% and AST levels by 44%, while also lowering hepatic TG by 22% and TC by 15%. Transcriptome analysis revealed that ME ameliorated NAFLD through AMPK/PPAR-γ/NF-κB axis. Metabolomic analysis demonstrated the involvement of unsaturated fatty acid and steroid hormone biosynthesis in NAFLD metabolism. CONCLUSIONS Our study demonstrates that ME alleviates NAFLD progression by regulating the AMPK/PPAR-γ/NF-κB signaling axis. However, these findings are based on preclinical animal studies, and further investigation is required to determine the clinical applicability of these effects in human patients.
Collapse
Affiliation(s)
- Li-Juan Zhang
- Xinjiang Academy of Forestry, Key Laboratory of Forest Resources and Utilization in Xinjiang of National Forestry and Grassland Administration, Urumqi, Xinjiang 830000, China; Pharmaceutics College of Shihezi University/Key Laboratory of Xinjiang Plant Drug Resources Utilization Ministry of Education, Shihezi, Xinjiang 832000, China
| | - Shun-Ying Li
- Xinjiang Academy of Forestry, Key Laboratory of Forest Resources and Utilization in Xinjiang of National Forestry and Grassland Administration, Urumqi, Xinjiang 830000, China; Pharmaceutics College of Shihezi University/Key Laboratory of Xinjiang Plant Drug Resources Utilization Ministry of Education, Shihezi, Xinjiang 832000, China
| | - Shao-Li Fan
- Xinjiang Academy of Forestry, Key Laboratory of Forest Resources and Utilization in Xinjiang of National Forestry and Grassland Administration, Urumqi, Xinjiang 830000, China
| | - Aobulikasimu Nuerbiye
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Jiu-Yang Zhao
- Xinjiang Academy of Forestry, Key Laboratory of Forest Resources and Utilization in Xinjiang of National Forestry and Grassland Administration, Urumqi, Xinjiang 830000, China
| | - Bahetiyaer Keremu
- Xinjiang Academy of Forestry, Key Laboratory of Forest Resources and Utilization in Xinjiang of National Forestry and Grassland Administration, Urumqi, Xinjiang 830000, China
| | - Lu Yang
- Xinjiang Academy of Forestry, Key Laboratory of Forest Resources and Utilization in Xinjiang of National Forestry and Grassland Administration, Urumqi, Xinjiang 830000, China.
| | - Ke Zhang
- Pharmaceutics College of Shihezi University/Key Laboratory of Xinjiang Plant Drug Resources Utilization Ministry of Education, Shihezi, Xinjiang 832000, China.
| | - Hang-Yu Wang
- Pharmaceutics College of Shihezi University/Key Laboratory of Xinjiang Plant Drug Resources Utilization Ministry of Education, Shihezi, Xinjiang 832000, China
| | - Jin-Hui Wang
- Xinjiang Academy of Forestry, Key Laboratory of Forest Resources and Utilization in Xinjiang of National Forestry and Grassland Administration, Urumqi, Xinjiang 830000, China; College of Pharmacy, Harbin Medical University, Harbin 150081, Heilongjiang, China
| |
Collapse
|
2
|
Lv M, Liu C, Jiang X, Ge M, Wang H, Yu W. Molting of laying hens can activate AMPK- lipophagy - lipid metabolism pathway and improve intestinal digestion and absorption. Poult Sci 2025; 104:104641. [PMID: 39667182 PMCID: PMC11699241 DOI: 10.1016/j.psj.2024.104641] [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: 07/30/2024] [Revised: 11/30/2024] [Accepted: 12/04/2024] [Indexed: 12/14/2024] Open
Abstract
Poultry molting is a natural phenomenon, and process that improves the physiological function of laying hens. In this study, artificial intervention was used to induced molting (IM) in aged hens and improve their egg- laying performance. Jejunal lipophagy and lipid metabolism data were analyzed to elucidate the regulatory mechanisms by which the intestine affects egg production performance, particularly through the lens of digestion and absorption processes. Molting process in laying hens facilitated the regeneration of small intestinal villi following damage and shedding, while also reducing excess lipid accumulation within the intestine. Analyses of lipophagy and lipid metabolism-related factors revealed, increased the expression levels of genes and proteins, such as AMPK, FOXO1, TFEB, TFE3, PGC-1α and PPAR-α (P<0.05, P<0.01 and P<0.001). Serological analysis and detection of enzymes involved in digestion and absorption, showed upregulated expression of GLUT2, FABP (P<0.05 and P<0.001) and CD36 (P<0.01), and the activities of amylase, chymotrypsin and Lipase also increased significantly (P<0.05, P<0.01 and P<0.001). In conclusion, artificially IM activates the AMPK-lipophagy-lipid metabolism pathway to enhance intestinal digestion and absorption in laying hens. Our findings offer a theoretical framework for the intentional use of IM to promote a healthy state of digestion and absorption.
Collapse
Affiliation(s)
- Meiwei Lv
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Chenxin Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Xiaowen Jiang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Provincial Key Laboratory of Comparative Medicine and Animal Pathogenesis, Northeast Agricultural University, Harbin 150030, PR China
| | - Ming Ge
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Institute of Traditional Chinese Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Haibin Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Institute of Traditional Chinese Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Wenhui Yu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Institute of Traditional Chinese Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
3
|
Xu M, Chen ZY, Li Y, Li Y, Guo G, Dai RZ, Ni N, Tao J, Wang HY, Chen QL, Wang H, Zhou H, Yang YN, Chen S, Chen L. Rab2A-mediated Golgi-lipid droplet interactions support very-low-density lipoprotein secretion in hepatocytes. EMBO J 2024; 43:6383-6409. [PMID: 39496977 PMCID: PMC11649929 DOI: 10.1038/s44318-024-00288-x] [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: 04/28/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 11/06/2024] Open
Abstract
Lipid droplets (LDs) serve as crucial hubs for lipid trafficking and metabolic regulation through their numerous interactions with various organelles. While the interplay between LDs and the Golgi apparatus has been recognized, their roles and underlying mechanisms remain poorly understood. Here, we reveal the role of Ras-related protein Rab-2A (Rab2A) in mediating LD-Golgi interactions, thereby contributing to very-low-density lipoprotein (VLDL) lipidation and secretion in hepatocytes. Mechanistically, our findings identify a selective interaction between Golgi-localized Rab2A and 17-beta-hydroxysteroid dehydrogenase 13 (HSD17B13) protein residing on LDs. This complex facilitates dynamic organelle communication between the Golgi apparatus and LDs, thus contributing to lipid transfer from LDs to the Golgi apparatus for VLDL2 lipidation and secretion. Attenuation of Rab2A activity via AMP-activated protein kinase (AMPK) suppresses the Rab2A-HSD17B13 complex formation, impairing LD-Golgi interactions and subsequent VLDL secretion. Furthermore, genetic inhibition of Rab2A and HSD17B13 in the liver reduces the serum triglyceride and cholesterol levels. Collectively, this study provides a new perspective on the interactions between the Golgi apparatus and LDs.
Collapse
Affiliation(s)
- Min Xu
- College of Life Sciences, Anhui Medical University, 230032, Hefei, China
| | - Zi-Yue Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, 210061, Nanjing, China
| | - Yang Li
- Department of Cardiology, People's Hospital of Xinjiang Uyghur Autonomous Region, 830000, Urumqi, China
- Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, 830000, Urumqi, China
| | - Yue Li
- College of Life Sciences, Anhui Medical University, 230032, Hefei, China
| | - Ge Guo
- College of Life Sciences, Anhui Medical University, 230032, Hefei, China
| | - Rong-Zheng Dai
- College of Life Sciences, Anhui Medical University, 230032, Hefei, China
| | - Na Ni
- College of Life Sciences, Anhui Medical University, 230032, Hefei, China
| | - Jing Tao
- Department of Cardiology, People's Hospital of Xinjiang Uyghur Autonomous Region, 830000, Urumqi, China
- Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, 830000, Urumqi, China
| | - Hong-Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, 210061, Nanjing, China
| | - Qiao-Li Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, 210061, Nanjing, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, 230022, Hefei, China
| | - Hong Zhou
- College of Life Sciences, Anhui Medical University, 230032, Hefei, China.
| | - Yi-Ning Yang
- Department of Cardiology, People's Hospital of Xinjiang Uyghur Autonomous Region, 830000, Urumqi, China.
- Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, 830000, Urumqi, China.
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, 830000, Urumqi, China.
- Key Laboratory of Cardiovascular Disease Research, First Affiliated Hospital of Xinjiang Medical University, 830000, Urumqi, China.
| | - Shuai Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, 210061, Nanjing, China.
| | - Liang Chen
- College of Life Sciences, Anhui Medical University, 230032, Hefei, China.
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, 230001, Hefei, China.
| |
Collapse
|
4
|
Cao MM, Li YM, Ding X, Fang F, Yang LY. ARL8B promotes hepatocellular carcinoma progression and inhibits antitumor activity of lenvatinib via MAPK/ERK signaling by interacting with RAB2A. Cell Signal 2024; 124:111470. [PMID: 39413890 DOI: 10.1016/j.cellsig.2024.111470] [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: 07/01/2024] [Revised: 09/30/2024] [Accepted: 10/11/2024] [Indexed: 10/18/2024]
Abstract
Tumor recurrence and metastasis are important factors affecting postoperative survival in hepatocellular carcinoma (HCC) patients. ADP Ribosylation factor-like GTPase 8B (ARL8B) plays a crucial role in many biological processes, including lysosomal function, immune response, and cellular communication, all of which are related to the occurrence and development of tumors. However, its role in HCC remains unclear. Herein, we revealed that ARL8B is consistently elevated in HCC tissues compared to normal liver tissues, suggesting an unfavorable outcome in HCC patients. Increased ARL8B levels promoted the malignant phenotype of HCC in vitro and in vivo. Notably, ARL8B also induced epithelial-to-mesenchymal transition (EMT) in HCC cells. Mechanistically, the results of bioinformatics analysis combined with mass spectrometry revealed the potential downstream target molecule RAB2A of ARL8B. ARL8B directly interacted with RAB2A and increased the levels of GTP-bound RAB2A, thereby contributing to the activation of the extracellular signal-regulated kinase (ERK) signaling pathway. Interestingly, knockout of ARL8B in Hep3B cells enhanced the antitumor activity of lenvatinib in vitro and in vivo. Furthermore, AAV-shARL8B enhanced the inhibition of HCC growth through lenvatinib, providing new insights into its mechanism of action in lenvatinib-insensitive patients. In conclusion, ARL8B promotes the malignant phenotype of HCC and EMT via RAB2A mediated activation of the MAPK/ERK signaling pathway and is expected to be a valuable prognostic indicator and therapeutic target for HCC patients.
Collapse
Affiliation(s)
- Mo-Mo Cao
- Liver Cancer Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yi-Ming Li
- Liver Cancer Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiang Ding
- Department of Organ Transplantation, Xiangya Hospital, Central South University, Changsha, China
| | - Feng Fang
- Department of Hepatobiliary, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
| | - Lian-Yue Yang
- Liver Cancer Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
5
|
Lupi M, Avanzato D, Confalonieri S, Martino F, Pennisi R, Pupo E, Audrito V, Freddi S, Bertalot G, Montani F, Matoskova B, Sigismund S, Di Fiore PP, Lanzetti L. TBC1 domain-containing proteins are frequently involved in triple-negative breast cancers in connection with the induction of a glycolytic phenotype. Cell Death Dis 2024; 15:647. [PMID: 39231952 PMCID: PMC11375060 DOI: 10.1038/s41419-024-07037-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/06/2024]
Abstract
Metabolic plasticity is a hallmark of cancer, and metabolic alterations represent a promising therapeutic target. Since cellular metabolism is controlled by membrane traffic at multiple levels, we investigated the involvement of TBC1 domain-containing proteins (TBC1Ds) in the regulation of cancer metabolism. These proteins are characterized by the presence of a RAB-GAP domain, the TBC1 domain, and typically function as attenuators of RABs, the master switches of membrane traffic. However, a number of TBC1Ds harbor mutations in their catalytic residues, predicting biological functions different from direct regulation of RAB activities. Herein, we report that several genes encoding for TBC1Ds are expressed at higher levels in triple-negative breast cancers (TNBC) vs. other subtypes of breast cancers (BC), and predict prognosis. Orthogonal transcriptomics/metabolomics analysis revealed that the expression of prognostic TBC1Ds correlates with elevated glycolytic metabolism in BC cell lines. In-depth investigations of the three top hits from the previous analyses (TBC1D31, TBC1D22B and TBC1D7) revealed that their elevated expression is causal in determining a glycolytic phenotype in TNBC cell lines. We further showed that the impact of TBC1D7 on glycolytic metabolism of BC cells is independent of its known participation in the TSC1/TSC2 complex and consequent downregulation of mTORC1 activity. Since TBC1D7 behaves as an independent prognostic biomarker in TNBC, it could be used to distinguish good prognosis patients who could be spared aggressive therapy from those with a poor prognosis who might benefit from anti-glycolytic targeted therapies. Together, our results highlight how TBC1Ds connect disease aggressiveness with metabolic alterations in TNBC. Given the high level of heterogeneity among this BC subtype, TBC1Ds could represent important tools in predicting prognosis and guiding therapy decision-making.
Collapse
Grants
- IG #22811 Associazione Italiana per la Ricerca sul Cancro (Italian Association for Cancer Research)
- MFAG-2021 #26004 Associazione Italiana per la Ricerca sul Cancro (Italian Association for Cancer Research)
- IG #24415 Associazione Italiana per la Ricerca sul Cancro (Italian Association for Cancer Research)
- IG #23060 Associazione Italiana per la Ricerca sul Cancro (Italian Association for Cancer Research)
- PRIN 2020 Prot. 2020R2BP2E Ministero dell'Istruzione, dell'Università e della Ricerca (Ministry of Education, University and Research)
- PRIN 2022 Prot. 2022W93FTW Ministero dell'Istruzione, dell'Università e della Ricerca (Ministry of Education, University and Research)
- PRIN 2020 Prot. 2020R2BP2E Ministero dell'Istruzione, dell'Università e della Ricerca (Ministry of Education, University and Research)
- Ricerca Corrente 2023-2024 Ministero della Salute (Ministry of Health, Italy)
- 5x1000 Ministero della Salute (Ministry of Health, Italy)
- Ricerca Corrente 2023-2024 Ministero della Salute (Ministry of Health, Italy)
- 5x1000 Ministero della Salute (Ministry of Health, Italy)
- Ricerca Finalizzata RF-2021-12373957 Ministero della Salute (Ministry of Health, Italy)
- Ricerca Corrente 2023-2024 Ministero della Salute (Ministry of Health, Italy)
- 5x1000 Ministero della Salute (Ministry of Health, Italy)
Collapse
Affiliation(s)
- Mariadomenica Lupi
- Department of Oncology, University of Torino Medical School, Turin, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
| | - Daniele Avanzato
- Department of Oncology, University of Torino Medical School, Turin, Italy
- Department of Veterinary Sciences, Infectious Diseases Unit, University of Torino, Turin, Italy
| | | | - Flavia Martino
- Department of Oncology, University of Torino Medical School, Turin, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
| | - Rosa Pennisi
- Department of Oncology, University of Torino Medical School, Turin, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
| | | | - Valentina Audrito
- Department of Science and Technological Innovation (DISIT), University of Eastern Piedmont, Alessandria, Italy
| | - Stefano Freddi
- IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milan, Milan, Italy
| | - Giovanni Bertalot
- IEO, European Institute of Oncology IRCCS, Milan, Italy
- Unità Operativa Multizonale di Anatomia Patologica, APSS, Trento, Italy, and Centre for Medical Sciences - CISMed, University of Trento, Trento, Italy
| | | | | | - Sara Sigismund
- IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milan, Milan, Italy
| | - Pier Paolo Di Fiore
- IEO, European Institute of Oncology IRCCS, Milan, Italy.
- Department of Oncology and Haemato-Oncology, University of Milan, Milan, Italy.
| | - Letizia Lanzetti
- Department of Oncology, University of Torino Medical School, Turin, Italy.
- Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy.
| |
Collapse
|
6
|
Wang Q, Rong P, Zhang W, Yang X, Chen L, Cao Y, Liu M, Feng W, Ouyang Q, Chen Q, Li H, Liang H, Meng F, Wang HY, Chen S. TBC1D1 is an energy-responsive polarization regulator of macrophages via governing ROS production in obesity. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1899-1914. [PMID: 38902450 DOI: 10.1007/s11427-024-2628-1] [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: 04/25/2024] [Accepted: 05/23/2024] [Indexed: 06/22/2024]
Abstract
Energy status is linked to the production of reactive oxygen species (ROS) in macrophages, which is elevated in obesity. However, it is unclear how ROS production is upregulated in macrophages in response to energy overload for mediating the development of obesity. Here, we show that the Rab-GTPase activating protein (RabGAP) TBC1D1, a substrate of the energy sensor AMP-activated protein kinase (AMPK), is a critical regulator of macrophage ROS production and consequent adipose inflammation for obesity development. TBC1D1 deletion decreases, whereas an energy overload-mimetic non-phosphorylatable TBC1D1S231A mutation increases, ROS production and M1-like polarization in macrophages. Mechanistically, TBC1D1 and its downstream target Rab8a form an energy-responsive complex with NOX2 for ROS generation. Transplantation of TBC1D1S231A bone marrow aggravates diet-induced obesity whereas treatment with an ultra-stable TtSOD for removal of ROS selectively in macrophages alleviates both TBC1D1S231A mutation- and diet-induced obesity. Our findings therefore have implications for drug discovery to combat obesity.
Collapse
Affiliation(s)
- Qi Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Ping Rong
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Wen Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Xinyu Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Liang Chen
- College of Life Science, Anhui Medical University, Hefei, 230032, China
| | - Ye Cao
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Minjun Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Weikuan Feng
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Qian Ouyang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Qiaoli Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Hailong Li
- Redox Medical Center for Public Health, Medical College of Soochow University, Suzhou, 215123, China
| | - Hui Liang
- Department of General Surgery, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Fanguo Meng
- Redox Medical Center for Public Health, Medical College of Soochow University, Suzhou, 215123, China
| | - Hong-Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China.
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China.
| | - Shuai Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China.
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China.
| |
Collapse
|
7
|
Ashraf N, Van Nostrand JL. Fine-tuning AMPK in physiology and disease using point-mutant mouse models. Dis Model Mech 2024; 17:dmm050798. [PMID: 39136185 PMCID: PMC11340815 DOI: 10.1242/dmm.050798] [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] [Indexed: 08/17/2024] Open
Abstract
AMP-activated protein kinase (AMPK) is an evolutionarily conserved serine/threonine kinase that monitors the cellular energy status to adapt it to the fluctuating nutritional and environmental conditions in an organism. AMPK plays an integral part in a wide array of physiological processes, such as cell growth, autophagy and mitochondrial function, and is implicated in diverse diseases, including cancer, metabolic disorders, cardiovascular diseases and neurodegenerative diseases. AMPK orchestrates many different physiological outcomes by phosphorylating a broad range of downstream substrates. However, the importance of AMPK-mediated regulation of these substrates in vivo remains an ongoing area of investigation to better understand its precise role in cellular and metabolic homeostasis. Here, we provide a comprehensive overview of our understanding of the kinase function of AMPK in vivo, as uncovered from mouse models that harbor phosphorylation mutations in AMPK substrates. We discuss some of the inherent limitations of these mouse models, highlight the broader implications of these studies for understanding human health and disease, and explore the valuable insights gained that could inform future therapeutic strategies for the treatment of metabolic and non-metabolic disorders.
Collapse
Affiliation(s)
- Naghmana Ashraf
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jeanine L. Van Nostrand
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
8
|
Mohammed DM, Salem MB, Elzallat M, Hammam OA, Suliman AA. Moringa oleifera L. mediated zinc oxide nano-biofertilizer alleviates non-alcoholic steatohepatitis via modulating de novo lipogenesis pathway and miRNA-122 expression. FOOD BIOSCI 2024; 60:104286. [DOI: 10.1016/j.fbio.2024.104286] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2024]
|
9
|
Lu C, Liu D, Li M, Shi X, Guan J, Song G, Yin Y, Zheng M, Ma F, Liu G. GPR30 selective agonist G-1 induced insulin resistance in ovariectomized mice on high fat diet and its mechanism. Biochem Biophys Res Commun 2024; 716:150026. [PMID: 38701557 DOI: 10.1016/j.bbrc.2024.150026] [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: 02/23/2024] [Revised: 04/19/2024] [Accepted: 04/26/2024] [Indexed: 05/05/2024]
Abstract
BACKGROUND Previous in vivo and in vitro studies have demonstrated that estrogen receptor agonist G-1 regulates glucose and lipid metabolism. This study focused on the effects of G-1 on cardiometabolic syndrome and anti-obesity under a high fat diet (HFD). METHODS Bilateral ovariectomized female mice were fed an HFD for 6 weeks, and treated them with G-1. A cardiomyocyte insulin resistance model was used to simulate the in vivo environment. The main outcome measures were blood glucose, body weight, and serum insulin levels to assess insulin resistance, while cardiac function and degree of fibrosis were assessed by cardiac ultrasound and pathological observations. We also examined the expression of p-AMPK, p-AKT, and GLUT4 in mice hearts and in vitro models to explore the mechanism by which G-1 regulates insulin signaling. RESULTS G-1 reduced body weight in mice on an HFD, but simultaneously increased blood glucose and promoted insulin resistance, resulting in myocardial damage. This damage included disordered cardiomyocytes, massive accumulation of glycogen, extensive fibrosis of the heart, and thickening of the front and rear walls of the left ventricle. At the molecular level, G-1 enhances gluconeogenesis and promotes glucose production by increasing the activity of pyruvate carboxylase (PC) while inhibiting GLUT4 translocation via the AMPK/TBC1D1 pathway, thereby limiting glucose uptake. CONCLUSION Despite G-1's the potential efficacy in weight reduction, the concomitant induction of insulin resistance and cardiac impairment in conjunction with an HFD raises significant concerns. Therefore, comprehensive studies of its safety profile and effects under specific conditions are essential prior to clinical use.
Collapse
Affiliation(s)
- Congcong Lu
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, 050000, Hebei, China
| | - Da Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, 050000, Hebei, China
| | - Min Li
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, 050000, Hebei, China
| | - Xiaocui Shi
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, 050000, Hebei, China
| | - Jingyue Guan
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, 050000, Hebei, China
| | - Guoyuan Song
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, 050000, Hebei, China
| | - Yajuan Yin
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, 050000, Hebei, China
| | - Mingqi Zheng
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, 050000, Hebei, China
| | - Fangfang Ma
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, 050000, Hebei, China.
| | - Gang Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, 050000, Hebei, China; Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang, Hebei, China; Hebei Key Laboratory of Heart and Metabolism, Shijiazhuang, Hebei, China; Hebei Engineering Research Center of Intelligent Medical Clinical Application, Shijiazhuang, Hebei, China; Hebei International Joint Research Center for Structural Heart Disease, Shijiazhuang, Hebei, China.
| |
Collapse
|
10
|
Zhang P, Zhu L, Pan X. A comprehensive analysis of the oncogenic and prognostic role of TBC1Ds in human hepatocellular carcinoma. PeerJ 2024; 12:e17362. [PMID: 38766486 PMCID: PMC11100476 DOI: 10.7717/peerj.17362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 04/18/2024] [Indexed: 05/22/2024] Open
Abstract
Backgrounds TBC1D family members (TBC1Ds) are a group of proteins that contain the Tre2-Bub2-Cdc16 (TBC) domain. Recent studies have shown that TBC1Ds are involved in tumor growth, but no analysis has been done of expression patterns and prognostic values of TBC1Ds in hepatocellular carcinoma (HCC). Methods The expression levels of TBC1Ds were evaluated in HCC using the TIMER, UALCN and Protein Atlas databases. The correlation between the mRNA levels of TBC1Ds and the prognosis of patients with HCC in the GEPIA database was then analyzed. An enrichment analysis then revealed genes that potentially interact with TBC1Ds. The correlation between levels of TBC1Ds and tumor-infiltrating immune cells (TIICs) in HCC were studied using the TIMER 2.0 database. Finally, a series of in vitro assays verified the role of TBC1Ds in HCC progression. Results This study revealed the upregulated expression of TBC1Ds in HCC and the strong positive correlation between the mRNA levels of TBC1Ds and poor prognosis of patients with HCC. The functions of TBC1Ds were mainly related to autophagy and the AMPK pathway. There was also a significant correlation between level of TBC1Ds and tumor-infiltrating immune cells (TIICs) in HCC. The promoting role of TBC1Ds in HCC progression was verified in vitro assays. Conclusion The results of this analysis indicate that TBC1Ds may serve as new biomarkers for early diagnosis and treatment of HCC.
Collapse
MESH Headings
- Humans
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/metabolism
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Liver Neoplasms/immunology
- Liver Neoplasms/metabolism
- Prognosis
- GTPase-Activating Proteins/genetics
- GTPase-Activating Proteins/metabolism
- Gene Expression Regulation, Neoplastic
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Autophagy/genetics
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Cell Line, Tumor
Collapse
Affiliation(s)
- Pei Zhang
- The Affiliated Geriatric Hospital of Nanjing Medical University, Nanjing, China
| | - Lei Zhu
- The Affiliated Geriatric Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaodong Pan
- The Affiliated Geriatric Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
11
|
Lv T, Lou Y, Yan Q, Nie L, Cheng Z, Zhou X. Phosphorylation: new star of pathogenesis and treatment in steatotic liver disease. Lipids Health Dis 2024; 23:50. [PMID: 38368351 PMCID: PMC10873984 DOI: 10.1186/s12944-024-02037-9] [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: 09/26/2023] [Accepted: 01/31/2024] [Indexed: 02/19/2024] Open
Abstract
Steatotic liver disease poses a serious threat to human health and has emerged as one of the most significant burdens of chronic liver disease worldwide. Currently, the research mechanism is not clear, and there is no specific targeted drug for direct treatment. Phosphorylation is widely regarded as the most common type of protein modification, closely linked to steatotic liver disease in previous studies. However, there is no systematic review to clarify the relationship and investigate from the perspective of phosphorylation. Phosphorylation has been found to mainly regulate molecule stability, affect localization, transform molecular function, and cooperate with other protein modifications. Among them, adenosine 5'-monophosphate-activated protein kinase (AMPK), serine/threonine kinase (AKT), and nuclear factor kappa-B (NF-kB) are considered the core mechanisms in steatotic liver disease. As to treatment, lifestyle changes, prescription drugs, and herbal ingredients can alleviate symptoms by influencing phosphorylation. It demonstrates the significant role of phosphorylation as a mechanism occurrence and a therapeutic target in steatotic liver disease, which could be a new star for future exploration.
Collapse
Affiliation(s)
- Tiansu Lv
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yan Lou
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Qianhua Yan
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lijuan Nie
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhe Cheng
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiqiao Zhou
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
12
|
Marcondes-de-Castro IA, Reis-Barbosa PH, Marinho TS, Aguila MB, Mandarim-de-Lacerda CA. AMPK/mTOR pathway significance in healthy liver and non-alcoholic fatty liver disease and its progression. J Gastroenterol Hepatol 2023; 38:1868-1876. [PMID: 37438882 DOI: 10.1111/jgh.16272] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/06/2023] [Accepted: 06/14/2023] [Indexed: 07/14/2023]
Abstract
Obesity is related to several organs, but the liver is particularly affected. Adenosine monophosphate-activated protein kinase (AMPK) is a cellular energy sensor and regulator of liver lipid dysfunction and glucose metabolism. The mechanistic target of rapamycin (mTOR) is a protein kinase regulating cell growth, survival, metabolism, and immunity. Together, these pathways are involved in obesity, insulin resistance, non-alcoholic fatty liver disease (NAFLD) and its progression, and autophagy. During energy demand, liver kinase B (LKB) phosphorylation helps activate the AMPK/mTOR pathways. Likewise, the protein forkhead box O family (FOXO) negatively regulates adipogenesis by binding to the promoter sites of peroxisome proliferator-activated receptor-gamma coactivator 1-alpha, initiating adipogenesis. In addition, acetyl-CoA carboxylase, which regulates de novo lipogenesis, is linked to LKB and FOXO in developing NAFLD. The kinase complex, consisting of Unc-51-like autophagy-activating kinase 1 or 2 (ULK1, ULK2) by stimulating autophagy, and eliminating fat droplets in NAFLD, is regulated by mTORC1 and negatively regulated by AMPK that suppresses liver lipogenesis and increases fatty acid oxidation. Also, ULK1 is essential for initiating phagophore formation, establishing macrophagy, and generating autophagosomes. The selective breakdown of lipid droplets through macroautophagy, or macrolipophagy, occurs on a cellular energy level using free fatty acids. In addition, mTORC1 promotes lipogenesis by activating sterol regulatory element-binding protein. Finding new components and novel regulatory modes in signaling is significant for a better understanding of the AMPK/mTOR pathways, potentially facilitating the development of future diagnostic and therapeutic strategies for NAFLD and its progression to non-alcoholic steatohepatitis, cirrhosis, and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Ilitch Aquino Marcondes-de-Castro
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Centre, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Pedro Henrique Reis-Barbosa
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Centre, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Thatiany Souza Marinho
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Centre, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Centre, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Carlos Alberto Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Centre, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| |
Collapse
|
13
|
Liu GT, Ni QF, Zhang YH, Dong XM, Zhou C, Shen B, Zhu JY, Chen YJ, Zhu Z. Application of noninvasive test (acoustic attenuation imaging and ultrasonic shear wave elastography) to grade nonalcoholic fatty liver disease: An observational study. Medicine (Baltimore) 2023; 102:e34550. [PMID: 37565905 PMCID: PMC10419497 DOI: 10.1097/md.0000000000034550] [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: 04/15/2023] [Accepted: 07/12/2023] [Indexed: 08/12/2023] Open
Abstract
The aim of this study was to validate the diagnostic efficacy of acoustic attenuation imaging (ATI) and ultrasonic shear wave elastography (SWE) in classifying nonalcoholic fatty liver disease (NAFLD). A total of 100 patients with NAFLD were recruited from our hospital between January 2021 and December 2022. Patient demographics and clinical data were collected, and 2-dimensional ultrasound was used to screen patients based on liver echo characteristics. Patients without liver space-occupying lesions underwent routine ultrasound examinations. Imaging or serology was used to confirm the presence of fatty liver in patients or healthy individuals. Patients with alcoholic liver disease (alcohol equivalent content < 20 g/day for women, <30 g/day for men), as well as those with lenticular degeneration, total parenteral nutrition, autoimmune liver disease, drug-induced hepatitis, and viral hepatitis, were excluded from the study. Out of the 100 included patients, 24 had normal liver, 21 had mild fatty liver, 30 had moderate fatty liver, and 25 had severe fatty liver. There were age differences between the normal group and patients with mild fatty liver, and the average body mass index (BMI) varied across the 4 groups. As the severity of the disease increased, the average BMI also increased (P < .05). The ATI scores and SWE scores differed significantly among the different groups (P < .05), with both scores showing an upward trend as the fatty liver condition worsened. Correlation analysis revealed positive correlations between ATI and SWE scores and the degree of fatty liver (P < .05), positive correlations with BMI (P < .05), and negative correlations with high-density lipoprotein cholesterol expression (P < .05). The area under the curve (AUC) for the ATI score in diagnosing different degrees of fatty liver was > 0.750, and the AUC for the SWE score was also > 0.750. The AUC for SWE score in diagnosing different degrees of fatty liver ranged from 1.01 to 4.57, while the combined AUC for ATI and SWE scores was > 0.850, with respective cutoff values of 3.62, 5.72, and 7.57 based on the maximum approximate entry index. The combination of ATI and SWE has a significant impact on the grading diagnosis of NAFLD, and its application can be extended to clinical practice.
Collapse
Affiliation(s)
- Guo Tao Liu
- Department of Ultrasonography, Taicang Affiliated Hospital of Soochow University (The First People’s Hospital of Taicang), Suzhou City, Jiangsu Province, China
| | - Qing Feng Ni
- Department of Ultrasonography, Taicang Affiliated Hospital of Soochow University (The First People’s Hospital of Taicang), Suzhou City, Jiangsu Province, China
| | - Yu Hang Zhang
- Department of Ultrasonography, Taicang Affiliated Hospital of Soochow University (The First People’s Hospital of Taicang), Suzhou City, Jiangsu Province, China
| | - Xue Ming Dong
- Department of Ultrasonography, Taicang Affiliated Hospital of Soochow University (The First People’s Hospital of Taicang), Suzhou City, Jiangsu Province, China
| | - Chao Zhou
- Department of Ultrasonography, Taicang Affiliated Hospital of Soochow University (The First People’s Hospital of Taicang), Suzhou City, Jiangsu Province, China
| | - Bo Shen
- Department of Ultrasonography, Taicang Affiliated Hospital of Soochow University (The First People’s Hospital of Taicang), Suzhou City, Jiangsu Province, China
| | - Jing Ya Zhu
- Department of Ultrasonography, Taicang Affiliated Hospital of Soochow University (The First People’s Hospital of Taicang), Suzhou City, Jiangsu Province, China
| | - Ya Juan Chen
- Department of Ultrasonography, Taicang Affiliated Hospital of Soochow University (The First People’s Hospital of Taicang), Suzhou City, Jiangsu Province, China
| | - Zheng Zhu
- Department of Ultrasonography, Taicang Affiliated Hospital of Soochow University (The First People’s Hospital of Taicang), Suzhou City, Jiangsu Province, China
| |
Collapse
|
14
|
Lee SM, Muratalla J, Sierra-Cruz M, Cordoba-Chacon J. Role of hepatic peroxisome proliferator-activated receptor γ in non-alcoholic fatty liver disease. J Endocrinol 2023; 257:e220155. [PMID: 36688873 PMCID: PMC10048618 DOI: 10.1530/joe-22-0155] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 01/23/2023] [Indexed: 01/24/2023]
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) belongs to a family of nuclear receptors that could serve as lipid sensors. PPARγ is the target of a group of insulin sensitizers called thiazolidinediones (TZDs) which regulate the expression of genes involved in glucose and lipid metabolism as well as adipokines that regulate metabolic function in other tissues. Non-alcoholic fatty liver disease (NAFLD) has a high prevalence worldwide and is even higher in patients with obesity and insulin resistance. TZD-mediated activation of PPARγ could serve as a good treatment for NAFLD because TZDs have shown anti-fibrogenic and anti-inflammatory effectsin vitro and increase insulin sensitivity in peripheral tissues which improves liver pathology. However, mechanistic studies in mouse models suggest that the activation of PPARγ in hepatocytes might reduce or limit the therapeutic potential of TZD against NAFLD. In this review, we briefly describe the short history of PPAR isoforms, the relevance of their expression in different tissues, as well as the pathogenesis and potential therapeutics for NAFLD. We also discuss some evidence derived from mouse models that could be useful for endocrinologists to assess tissue-specific roles of PPARs, complement reverse endocrinology approaches, and understand the direct role that PPARγ has in hepatocytes and non-parenchymal cells.
Collapse
Affiliation(s)
- Samuel M. Lee
- Department of Medicine. Division of Endocrinology, Diabetes and Metabolism. University of Illinois at Chicago, Chicago. IL
| | - Jose Muratalla
- Department of Medicine. Division of Endocrinology, Diabetes and Metabolism. University of Illinois at Chicago, Chicago. IL
| | - Marta Sierra-Cruz
- Department of Medicine. Division of Endocrinology, Diabetes and Metabolism. University of Illinois at Chicago, Chicago. IL
| | - Jose Cordoba-Chacon
- Department of Medicine. Division of Endocrinology, Diabetes and Metabolism. University of Illinois at Chicago, Chicago. IL
| |
Collapse
|
15
|
Li J, Wu N, Yang Y, Zhai X, Yuan F, Zhang F, Yu N, Li D, Wang R, Wang J, Zhang L, Shi Y, He G, Liu B. Unique genetic variants of lean nonalcoholic fatty liver disease: a retrospective cohort study. BMC Endocr Disord 2023; 23:11. [PMID: 36627697 PMCID: PMC9830772 DOI: 10.1186/s12902-022-01234-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 11/30/2022] [Indexed: 01/12/2023] Open
Abstract
We investigated the prevalence and clinical metabolic characteristics of lean nonalcoholic fatty liver disease (NAFLD) in an elderly Chinese population and assessed the relevance of lipid markers and genetic variation. All 5,338 community subjects underwent detailed clinical and laboratory examinations and were divided into three groups: lean (Body mass index (BMI) < 23 kg/m2, n = 2,012), overweight (BMI = 23-24.9 kg/m2, n = 1,354), and obese (BMI ≥ 25 kg/m2, n = 1,972). Single nucleotide polymorphisms were selected based on those reported in previous NAFLD or obesity genome-wide association studies. The frequencies of alleles and genotypes were calculated and statistically analyzed with Pearson's χ2 tests. One-way ANCOVA was used to test the association between positive SNPs and metabolic parameters in lean NAFLD individuals. Our results showed that the C allele frequency of rs2279026, the G allele of rs2279028, the C allele of rs780093, and the C allele frequency of rs1260326 were higher in obese NAFLD than in lean NAFLD (P < 0.05). In addition, we observed an association between the CC of rs1421085, TT of rs3751812, AA of rs8050136, and AA of rs9939609 genotypes in the FTO gene and low-density lipoprotein levels (P < 0.05). In conclusion, our findings provide a unique perspective on the prevalence, genetic characteristics, and metabolic profile of NAFLD in older lean individuals in China. This is the first study to examine the association between genetic variants in the FTO, TFAP2B and GCKR genes and NAFLD in a cohort of lean individuals.
Collapse
Affiliation(s)
- Jie Li
- Shanghai Innovation Center of Traditional Chinese Medicine Health Service, School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Na Wu
- Shanghai Innovation Center of Traditional Chinese Medicine Health Service, School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China
| | - Yukun Yang
- Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Xiangyu Zhai
- Graduate School of Sport Sciences, Waseda University, Saitama, Japan
| | - Fan Yuan
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China
| | - Fengwei Zhang
- Shanghai Innovation Center of Traditional Chinese Medicine Health Service, School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ning Yu
- Shanghai Innovation Center of Traditional Chinese Medicine Health Service, School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dong Li
- Zhangjiang Community Health Service Center of Pudong New District, Shanghai, China
| | - Ruirui Wang
- Shanghai Innovation Center of Traditional Chinese Medicine Health Service, School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianying Wang
- Shanghai Innovation Center of Traditional Chinese Medicine Health Service, School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lei Zhang
- Shanghai Innovation Center of Traditional Chinese Medicine Health Service, School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yi Shi
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China
| | - Guang He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China.
| | - Baocheng Liu
- Shanghai Innovation Center of Traditional Chinese Medicine Health Service, School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
16
|
Lin D, Song Y. Dapagliflozin Presented Nonalcoholic Fatty Liver Through Metabolite Extraction and AMPK/NLRP3 Signaling Pathway. Horm Metab Res 2023; 55:75-84. [PMID: 36495240 DOI: 10.1055/a-1970-3388] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In recent years, the incidence rate of nonalcoholic fatty liver disease (NAFLD) has been increasing year by year. The experiments conducted on rat elucidated the effect and underlying mechanism of dapagliflozin in NAFLD. Sprague Dawley rats were fed with HFD (Fat accounts for 52%, carbohydrate 34% and protein 14%) for 12 weeks as NAFLD model. Dapagliflozin presented NAFLD in rat model. Dapagliflozin reduced oxidative stress and inflammation in rat model of NAFLD. Dapagliflozin reduced oxidative stress and inflammation in vitro model of NAFLD. Dapagliflozin in a model of NAFLD metabolized into histamine H1 receptor, caffeine metabolism, mannose type O-glycan biosynthesis, choline metabolism in cancer, tryptophan metabolism, and glycerophospholipid metabolism. Dapagliflozin induced AMPK/NLRP3 signaling pathway. The regulation of AMPK/NLRP3 signaling pathway affected the effects of dapagliflozin on nonalcoholic fatty liver. In summary, dapagliflozin plays a preventative role in NAFLD through metabolite extraction, the inhibition of oxidative stress, and inflammation by AMPK/NLRP3 signaling pathway. Dapagliflozin may be a potential therapeutic agent for oxidative stress and inflammation in model of NAFLD.
Collapse
Affiliation(s)
- Deng Lin
- Department of Endocrinology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Yuling Song
- Department of Endocrinology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
17
|
Fang X, Wang H, Tan X, Ye T, Xu Y, Fan J. Elevated Serum Regulator of Calcineurin 2 is Associated With an Increased Risk of Non-Alcoholic Fatty Liver Disease. Front Pharmacol 2022; 13:840764. [PMID: 35370729 PMCID: PMC8967172 DOI: 10.3389/fphar.2022.840764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/24/2022] [Indexed: 11/13/2022] Open
Abstract
Background: The promoting effect of the regulator of calcineurin 2 (RCAN2) in hepatic steatosis has been observed in animal studies. However, the association of RCAN2 with non-alcoholic fatty liver disease (NAFLD) in humans remains unclear. This study aimed to evaluate the expression of RCAN2 in the liver of mice with hepatic steatosis and in the serum of NAFLD patients and to explore the relationship between serum RCAN2 levels and NAFLD. Methods: The mRNA and protein expression of RCAN2 were detected by quantitative real-time PCR (qRT-PCR) and Western blot. NAFLD was diagnosed by abdominal ultrasonography. Circulating RCAN2 levels were measured by ELISA kits. The relationship between serum RCAN2 levels and NAFLD was assessed. Results: qRT-PCR and Western blot analysis showed that compared with the corresponding controls, the mRNA and protein expression of RCAN2 were significantly increased in the liver tissues of db/db and mice on a high-fat diet. Serum RCAN2 levels were markedly elevated in NAFLD patients compared with non-NAFLD subjects. Binary logistic regression analysis showed that serum RCAN2 levels were significantly associated with NAFLD. Receiver operation characteristic (ROC) curve analysis showed that serum RCAN2 might act as a predictive biomarker for NAFLD [area under the curve (AUC) = 0.663, 95% CI = 0.623–0.702], and the serum RCAN2/(AST/ALT) ratio displayed improved predictive accuracy (AUC = 0.816, 95% CI = 0.785–0.846). Conclusion: Elevated serum RCAN2 levels were associated with an increased risk of NAFLD. Serum RCAN2, especially the serum RCAN2/(AST/ALT) ratio, might be a candidate diagnostic marker for NAFLD.
Collapse
Affiliation(s)
- Xia Fang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, China.,Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China
| | - Hongya Wang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, China.,Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China
| | - Xiaozhen Tan
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, China.,Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China
| | - Ting Ye
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yong Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, China.,Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China
| | - Jiahao Fan
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|