201
|
Puengel T, Liu H, Guillot A, Heymann F, Tacke F, Peiseler M. Nuclear Receptors Linking Metabolism, Inflammation, and Fibrosis in Nonalcoholic Fatty Liver Disease. Int J Mol Sci 2022; 23:ijms23052668. [PMID: 35269812 PMCID: PMC8910763 DOI: 10.3390/ijms23052668] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/23/2022] [Accepted: 02/26/2022] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) and its progressive form nonalcoholic steatohepatitis (NASH) comprise a spectrum of chronic liver diseases in the global population that can lead to end-stage liver disease and hepatocellular carcinoma (HCC). NAFLD is closely linked to the metabolic syndrome, and comorbidities such as type 2 diabetes, obesity and insulin resistance aggravate liver disease, while NAFLD promotes cardiovascular risk in affected patients. The pathomechanisms of NAFLD are multifaceted, combining hepatic factors including lipotoxicity, mechanisms of cell death and liver inflammation with extrahepatic factors including metabolic disturbance and dysbiosis. Nuclear receptors (NRs) are a family of ligand-controlled transcription factors that regulate glucose, fat and cholesterol homeostasis and modulate innate immune cell functions, including liver macrophages. In parallel with metabolic derangement in NAFLD, altered NR signaling is frequently observed and might be involved in the pathogenesis. Therapeutically, clinical data indicate that single drug targets thus far have been insufficient for reaching patient-relevant endpoints. Therefore, combinatorial treatment strategies with multiple drug targets or drugs with multiple mechanisms of actions could possibly bring advantages, by providing a more holistic therapeutic approach. In this context, peroxisome proliferator-activated receptors (PPARs) and other NRs are of great interest as they are involved in wide-ranging and multi-organ activities associated with NASH progression or regression. In this review, we summarize recent advances in understanding the pathogenesis of NAFLD, focusing on mechanisms of cell death, immunometabolism and the role of NRs. We outline novel therapeutic strategies and discuss remaining challenges.
Collapse
Affiliation(s)
- Tobias Puengel
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, 13353 Berlin, Germany; (T.P.); (H.L.); (A.G.); (F.H.)
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
| | - Hanyang Liu
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, 13353 Berlin, Germany; (T.P.); (H.L.); (A.G.); (F.H.)
| | - Adrien Guillot
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, 13353 Berlin, Germany; (T.P.); (H.L.); (A.G.); (F.H.)
| | - Felix Heymann
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, 13353 Berlin, Germany; (T.P.); (H.L.); (A.G.); (F.H.)
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, 13353 Berlin, Germany; (T.P.); (H.L.); (A.G.); (F.H.)
- Correspondence: (F.T.); (M.P.)
| | - Moritz Peiseler
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, 13353 Berlin, Germany; (T.P.); (H.L.); (A.G.); (F.H.)
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
- Correspondence: (F.T.); (M.P.)
| |
Collapse
|
202
|
Mice with a deficiency in Peroxisomal Membrane Protein 4 (PXMP4) display mild changes in hepatic lipid metabolism. Sci Rep 2022; 12:2512. [PMID: 35169201 PMCID: PMC8847483 DOI: 10.1038/s41598-022-06479-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/31/2022] [Indexed: 11/08/2022] Open
Abstract
Peroxisomes play an important role in the metabolism of a variety of biomolecules, including lipids and bile acids. Peroxisomal Membrane Protein 4 (PXMP4) is a ubiquitously expressed peroxisomal membrane protein that is transcriptionally regulated by peroxisome proliferator-activated receptor α (PPARα), but its function is still unknown. To investigate the physiological function of PXMP4, we generated a Pxmp4 knockout (Pxmp4-/-) mouse model using CRISPR/Cas9-mediated gene editing. Peroxisome function was studied under standard chow-fed conditions and after stimulation of peroxisomal activity using the PPARα ligand fenofibrate or by using phytol, a metabolite of chlorophyll that undergoes peroxisomal oxidation. Pxmp4-/- mice were viable, fertile, and displayed no changes in peroxisome numbers or morphology under standard conditions. Also, no differences were observed in the plasma levels of products from major peroxisomal pathways, including very long-chain fatty acids (VLCFAs), bile acids (BAs), and BA intermediates di- and trihydroxycholestanoic acid. Although elevated levels of the phytol metabolites phytanic and pristanic acid in Pxmp4-/- mice pointed towards an impairment in peroxisomal α-oxidation capacity, treatment of Pxmp4-/- mice with a phytol-enriched diet did not further increase phytanic/pristanic acid levels. Finally, lipidomic analysis revealed that loss of Pxmp4 decreased hepatic levels of the alkyldiacylglycerol class of neutral ether lipids, particularly those containing polyunsaturated fatty acids. Together, our data show that while PXMP4 is not critical for overall peroxisome function under the conditions tested, it may have a role in the metabolism of (ether)lipids.
Collapse
|
203
|
Zhou Z, Cai Z, Zhang C, Yang B, Chen L, He Y, Zhang L, Li Z. Design, synthesis, and biological evaluation of novel dual FFA1 and PPARδ agonists possessing phenoxyacetic acid scaffold. Bioorg Med Chem 2022; 56:116615. [PMID: 35051813 DOI: 10.1016/j.bmc.2022.116615] [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: 10/14/2021] [Revised: 12/24/2021] [Accepted: 01/06/2022] [Indexed: 11/11/2022]
Abstract
The free fatty acid receptor 1 (FFA1/GPR40) and peroxisome proliferator-activated receptor δ (PPARδ) have been widely considered as promising targets for type 2 diabetes mellitus (T2DM) due to their respective roles in promoting insulin secretion and improving insulin sensitivity. Hence, the dual FFA1/PPARδ agonists may exert synergistic effects by simultaneously activating FFA1 and PPARδ. The present study performed systematic exploration around previously reported FFA1 agonist 2-(2-fluoro-4-((2'-methyl-4'-(3-(methylsulfonyl)propoxy)-[1,1'-biphenyl]-3-yl)methoxy)phenoxy)acetic acid (lead compound), leading to the identification of a novel dual FFA1/PPARδ agonist 2-(2-fluoro-4-((3-(6-methoxynaphthalen-2-yl)benzyl)oxy)phenoxy)acetic acid (the optimal compound), which displayed high selectivity over PPARα and PPARγ. In addition, the docking study provided us with detailed binding modes of the optimal compound in FFA1 and PPARδ. Furthermore, the optimal compound exhibited greater glucose-lowering effects than lead compound, which might attribute to its synergistic effects by simultaneously modulating insulin secretion and resistance. Moreover, the optimal compound has an acceptable safety profile in the acute toxicity study at a high dose of 500 mg/kg Therefore, our results provided a novel dual FFA1/PPARδ agonist with excellent glucose-lowering effects in vivo.
Collapse
Affiliation(s)
- Zongtao Zhou
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Key Laboratory of New Drug Discovery and Evaluation, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Zongyu Cai
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Congzi Zhang
- Xianning Central Hospital, The First Affiliated Hospital of Hubei University Of Science And Technology, Xianning 437000, PR China
| | - Benhui Yang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Lianru Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yepu He
- Xianning Central Hospital, The First Affiliated Hospital of Hubei University Of Science And Technology, Xianning 437000, PR China
| | - Luyong Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Key Laboratory of New Drug Discovery and Evaluation, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Zheng Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Key Laboratory of New Drug Discovery and Evaluation, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; National Key Clinical Department (Clinical Pharmacy), The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510006, PR China.
| |
Collapse
|
204
|
Møllerhøj MB, Veidal SS, Thrane KT, Oró D, Overgaard A, Salinas CG, Madsen MR, Pfisterer L, Vyberg M, Simon E, Broermann A, Vrang N, Jelsing J, Feigh M, Hansen HH. Hepatoprotective effects of semaglutide, lanifibranor and dietary intervention in the GAN diet‐induced obese and biopsy‐confirmed mouse model of NASH. Clin Transl Sci 2022; 15:1167-1186. [PMID: 35143711 PMCID: PMC9099137 DOI: 10.1111/cts.13235] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/23/2021] [Accepted: 01/18/2022] [Indexed: 11/30/2022] Open
Abstract
Non‐alcoholic steatohepatitis (NASH) has emerged as a major challenge for public health because of high global prevalence and lack of evidence‐based therapies. Most animal models of NASH lack sufficient validation regarding disease progression and pharmacological treatment. The Gubra‐Amylin NASH (GAN) diet‐induced obese (DIO) mouse demonstrate clinical translatability with respect to disease etiology and hallmarks of NASH. This study aimed to evaluate disease progression and responsiveness to clinically effective interventions in GAN DIO‐NASH mice. Disease phenotyping was performed in male C57BL/6J mice fed the GAN diet high in fat, fructose, and cholesterol for 28–88 weeks. GAN DIO‐NASH mice with biopsy‐confirmed NASH and fibrosis received low‐caloric dietary intervention, semaglutide (30 nmol/kg/day, s.c.) or lanifibranor (30 mg/kg/day, p.o.) for 8 and 12 weeks, respectively. Within‐subject change in nonalcoholic fatty liver disease (NAFLD) Activity Score (NAS) and fibrosis stage was evaluated using automated deep learning‐based image analysis. GAN DIO‐NASH mice showed clear and reproducible progression in NASH, fibrosis stage, and tumor burden with high incidence of hepatocellular carcinoma. Consistent with clinical trial outcomes, semaglutide and lanifibranor improved NAS, whereas only lanifibranor induced regression in the fibrosis stage. Dietary intervention also demonstrated substantial benefits on metabolic outcomes and liver histology. Differential therapeutic efficacy of semaglutide, lanifibranor, and dietary intervention was supported by quantitative histology, RNA sequencing, and blood/liver biochemistry. In conclusion, the GAN DIO‐NASH mouse model recapitulates various histological stages of NASH and faithfully reproduces histological efficacy profiles of compounds in advanced clinical development for NASH. Collectively, these features highlight the utility of GAN DIO‐NASH mice in preclinical drug development.
Collapse
Affiliation(s)
| | | | | | - Denise Oró
- Gubra, Hørsholm Kongevej 11B, DK‐2970 Hørsholm Denmark
| | | | | | | | - Larissa Pfisterer
- Boehringer‐Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88397, Biberach an der Riss Germany
| | - Mogens Vyberg
- Center for RNA Medicine, Department of Clinical Medicine, Aalborg University Copenhagen Denmark
| | - Eric Simon
- Boehringer‐Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88397, Biberach an der Riss Germany
| | - Andre Broermann
- Boehringer‐Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88397, Biberach an der Riss Germany
| | - Niels Vrang
- Gubra, Hørsholm Kongevej 11B, DK‐2970 Hørsholm Denmark
| | - Jacob Jelsing
- Gubra, Hørsholm Kongevej 11B, DK‐2970 Hørsholm Denmark
| | - Michael Feigh
- Gubra, Hørsholm Kongevej 11B, DK‐2970 Hørsholm Denmark
| | | |
Collapse
|
205
|
Huang J, Ru G, Sun J, Sun L, Li Z. Elevated RIF1 participates in the epigenetic abnormalities of zygotes by regulating histone modifications on MuERV-L in obese mice. Mol Med 2022; 28:17. [PMID: 35123389 PMCID: PMC8818203 DOI: 10.1186/s10020-022-00446-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/26/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Maternal obesity impairs embryonic developmental potential and significantly increases the risks of metabolic disorders in offspring. However, the epigenetic transmission mechanism of maternal metabolic abnormalities is still poorly understood. METHODS We established an obesity model in female mice by high-fat diet (HFD) feeding. The effects of the HFD on the developmental potential of oocytes and embryos, the metabolic phenotype, and epigenetic modifications were investigated. The efficacy of metformin administration was assessed. Finally, the regulatory pathway of epigenetic remodeling during zygotic genome activation (ZGA) was explored. RESULTS Maternal HFD consumption significantly impaired glucose tolerance and increased the risk of metabolic disorders in F0 and F1 mice. Maternal HFD consumption also decreased embryonic developmental potential, increased reactive oxygen species (ROS) and γH2AX levels, and reduced the mitochondrial membrane potential (MMP) within oocytes, causing high levels of oxidative stress damage and DNA damage. Starting with this clue, we observed significantly increased RIF1 levels and shortened telomeres in obese mice. Moreover, significant abnormal DNA methylation and histone modification remodeling were observed during ZGA in obese mice, which may be coregulated by RIF1 and the ZGA marker gene MuERV-L. Metformin treatment reduced RIF1 levels, and partially improved ZGA activation status by rescuing epigenetic modification remodeling in oocytes and preimplantation embryos of obese mice. RIF1 knockdown experiments employing Trim-Away methods showed that RIF1 degradation altered the H3K4me3 and H3K9me3 enrichment and then triggered the MuERV-L transcriptional activation. Moreover, ChIP-seq data analysis of RIF1 knockouts also showed that RIF1 mediates the transcriptional regulation of MuERV-L by changing the enrichment of H3K4me3 and H3K9me3 rather than by altered DNA methylation. CONCLUSION Elevated RIF1 in oocytes caused by maternal obesity may mediate abnormal embryonic epigenetic remodeling and increase metabolic risk in offspring by regulating histone modifications on MuERV-L, which can be partially rescued by metformin treatment.
Collapse
Affiliation(s)
- Jiliang Huang
- Department of Reproductive Center, the First Affiliated Hospital of Shantou University Medical College, No. 57 Changping Road, Jinping District, Shantou, Guangdong, 515041, People's Republic of China
| | - Gaizhen Ru
- Department of Reproductive Center, the First Affiliated Hospital of Shantou University Medical College, No. 57 Changping Road, Jinping District, Shantou, Guangdong, 515041, People's Republic of China
| | - Jiajia Sun
- Department of Reproductive Center, the First Affiliated Hospital of Shantou University Medical College, No. 57 Changping Road, Jinping District, Shantou, Guangdong, 515041, People's Republic of China
| | - Luying Sun
- Department of Reproductive Center, the First Affiliated Hospital of Shantou University Medical College, No. 57 Changping Road, Jinping District, Shantou, Guangdong, 515041, People's Republic of China
| | - Zhiling Li
- Department of Reproductive Center, the First Affiliated Hospital of Shantou University Medical College, No. 57 Changping Road, Jinping District, Shantou, Guangdong, 515041, People's Republic of China
| |
Collapse
|
206
|
Zhu S, Wang W, Zhang J, Ji S, Jing Z, Chen YQ. Slc25a5 regulates adipogenesis by modulating ERK signaling in OP9 cells. Cell Mol Biol Lett 2022; 27:11. [PMID: 35109789 PMCID: PMC8903613 DOI: 10.1186/s11658-022-00314-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 01/17/2022] [Indexed: 01/09/2023] Open
Abstract
Background A comprehensive understanding of the molecular mechanisms of adipogenesis is a critically important strategy for identifying new targets for obesity intervention. Methods Transcriptomic and lipidomic approaches were used to explore the functional genes regulating adipogenic differentiation and their potential mechanism of action in OP9 cells and adipose-derived stem cells. Oil Red O staining was used to detect oil droplets in adipocytes. Results RNA sequencing (RNA-seq) showed that Slc25a5 expression was significantly upregulated in adipogenic differentiation. Depletion of Slc25a5 led to the suppressed expression of adipogenesis-related genes, reduced the accumulation of triglycerides, and inhibited PPARγ protein expression. Moreover, the knockdown of Slc25a5 resulted in significant reduction of oxidative phosphorylation (OXPHOS) protein expression (ATP5A1, CQCRC2, and MTCO1) and ATP production. The RNA-seq and real-time quantitative polymerase chain reaction (RT–qPCR) results suggested that adipogenic differentiation is possibly mediated by ERK1/2 phosphorylation, and this hypothesis was confirmed by intervention with PD98059 (an ERK 1/2 inhibitor). Conclusions This study indicates that Slc25a5 inhibits adipogenesis and might be a new therapeutic target for the treatment of obesity. Supplementary Information The online version contains supplementary material available at 10.1186/s11658-022-00314-y.
Collapse
Affiliation(s)
- Shenglong Zhu
- Wuxi School of Medicine, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, Jiangsu, China.,Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, China
| | - Wei Wang
- Wuxi School of Medicine, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, Jiangsu, China
| | - Jingwei Zhang
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Siyu Ji
- Wuxi School of Medicine, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, Jiangsu, China
| | - Zhe Jing
- Wuxi School of Medicine, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, Jiangsu, China
| | - Yong Q Chen
- Wuxi School of Medicine, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, Jiangsu, China. .,Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, China. .,School of Food Science and Technology, Jiangnan University, Wuxi, China.
| |
Collapse
|
207
|
Mao Y, Yiran Z, Sisi L, Huixi C, Xia L, Ting W, Guolian D, Xinmei L, Sheng J, Meng Y, Huang H. Advanced paternal age increased metabolic risks in mice offspring. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166355. [DOI: 10.1016/j.bbadis.2022.166355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/07/2022] [Accepted: 01/25/2022] [Indexed: 10/19/2022]
|
208
|
Luan ZL, Zhang C, Ming WH, Huang YZ, Guan YF, Zhang XY. Nuclear receptors in renal health and disease. EBioMedicine 2022; 76:103855. [PMID: 35123268 PMCID: PMC8819107 DOI: 10.1016/j.ebiom.2022.103855] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/31/2021] [Accepted: 01/18/2022] [Indexed: 02/07/2023] Open
Abstract
As a major social and economic burden for the healthcare system, kidney diseases contribute to the constant increase of worldwide deaths. A deeper understanding of the underlying mechanisms governing the etiology, development and progression of kidney diseases may help to identify potential therapeutic targets. As a superfamily of ligand-dependent transcription factors, nuclear receptors (NRs) are critical for the maintenance of normal renal function and their dysfunction is associated with a variety of kidney diseases. Increasing evidence suggests that ligands for NRs protect patients from renal ischemia/reperfusion (I/R) injury, drug-induced acute kidney injury (AKI), diabetic nephropathy (DN), renal fibrosis and kidney cancers. In the past decade, some breakthroughs have been made for the translation of NR ligands into clinical use. This review summarizes the current understanding of several important NRs in renal physiology and pathophysiology and discusses recent findings and applications of NR ligands in the management of kidney diseases.
Collapse
Affiliation(s)
- Zhi-Lin Luan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China; Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China; Dalian Key Laboratory for Nuclear Receptors in Major Metabolic Diseases, Dalian, Liaoning 116044, China
| | - Cong Zhang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Wen-Hua Ming
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Ying-Zhi Huang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - You-Fei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China; Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China; Dalian Key Laboratory for Nuclear Receptors in Major Metabolic Diseases, Dalian, Liaoning 116044, China.
| | - Xiao-Yan Zhang
- Health Science Center, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
209
|
Tao T, Deng P, Wang Y, Zhang X, Guo Y, Chen W, Qin J. Microengineered Multi-Organoid System from hiPSCs to Recapitulate Human Liver-Islet Axis in Normal and Type 2 Diabetes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103495. [PMID: 34951149 PMCID: PMC8844474 DOI: 10.1002/advs.202103495] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/21/2021] [Indexed: 05/07/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is a systematic multi-organ metabolic disease, which is characterized by the dynamic interplay among different organs. The increasing incidence of T2DM reflects an urgent need for the development of in vitro human-relevant models for disease study and drug therapy. Here, a new microfluidic multi-organoid system is developed that recapitulates the human liver-pancreatic islet axis in normal and disease states. The system contains two compartmentalized regions connected by a microchannel network, enabling 3D co-culture of human induced pluripotent stem cells (hiPSC)-derived liver and islet organoids for up to 30 days under circulatory perfusion conditions. The co-cultured liver and islet organoids exhibit favorable growth and improved tissue-specific functions. Transcriptional analyses reveal the activation of metabolically relevant signaling pathways in the co-cultured organoids. Notably, the co-culture system facilitates sensitive glucose-stimulated insulin secretion from islet organoids and increased glucose utilization in liver organoids by glucose tolerance tests. Both liver and islet organoids display mitochondrial dysfunction and decreased glucose transport capacity under high glucose conditions, which can be alleviated by metformin treatment. This novel multi-organoid system can recapitulate human-relevant liver-islet axis under both physiological and pathological conditions, providing a unique platform for future T2DM research and drug development.
Collapse
Affiliation(s)
- Tingting Tao
- Division of BiotechnologyDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
| | - Pengwei Deng
- Division of BiotechnologyDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yaqing Wang
- Division of BiotechnologyDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
| | - Xu Zhang
- Division of BiotechnologyDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
| | - Yaqiong Guo
- Division of BiotechnologyDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
| | - Wenwen Chen
- Division of BiotechnologyDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
| | - Jianhua Qin
- Division of BiotechnologyDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijing100101China
- CAS Center for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghai200031China
- University of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
210
|
Cariou B. The metabolic triad of non-alcoholic fatty liver disease, visceral adiposity and type 2 diabetes: Implications for treatment. Diabetes Obes Metab 2022; 24 Suppl 2:15-27. [PMID: 35014161 DOI: 10.1111/dom.14651] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 01/06/2022] [Accepted: 01/06/2022] [Indexed: 12/11/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is associated with visceral obesity, insulin resistance, type 2 diabetes (T2D) and has been often considered as the hepatic expression of the metabolic syndrome (MetS). Epidemiological studies highlight a bidirectional relationship of NAFLD with T2D in which NAFLD increases the risk of incident T2D and T2D increases the risk of severe non-alcoholic steatohepatitis (NASH) and liver fibrosis. Regarding the molecular determinants of NAFLD, we specifically focused in this review on adipocyte dysfunction as a key molecular link between visceral adipose tissue, MetS and NAFLD. Notably, the subcutaneous white adipose tissue expandability appears a critical adaptive buffering mechanism to prevent lipotoxicity and its related metabolic complications, such as NAFLD and T2D. There is a clinical challenge to consider therapeutic strategies targeting the metabolic dysfunction common to NASH and T2D pathogenesis. Strategies that promote significant and sustained weight loss (~10% of total body weight) such as metabolic and bariatric surgery or incretin-based therapies (GLP-1 receptor agonists or dual GLP-1/GIP or GLP-1/glucagon receptor co-agonists) are among the most efficient ones. In addition, insulin sensitizers such as PPARγ (pioglitazone) and pan-PPARs agonists (lanifibranor) have shown some beneficial effects on both NASH and liver fibrosis. Since NASH is a complex and multifactorial disease, it is conceivable that targeting different pathways, not only insulin resistance but also inflammation and fibrotic processes, is required to achieve NASH resolution.
Collapse
Affiliation(s)
- Bertrand Cariou
- Université de Nantes, Inserm, CNRS, CHU Nantes, l'institut du thorax, Nantes, France
| |
Collapse
|
211
|
Lan X, Han J, Wang B, Sun M. Integrated analysis of transcriptome profiling of lncRNAs and mRNAs in livers of type 2 diabetes mellitus. Physiol Genomics 2022; 54:86-97. [PMID: 35073196 DOI: 10.1152/physiolgenomics.00105.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) influence the progression of almost all human diseases, but the participation of lncRNAs in type 2 diabetes mellitus (T2DM) has not been fully elucidated. The present study aimed to systematically compare the transcriptome profiling of lncRNAs and mRNAs in livers between T2DM patients and controls, to identify key genes associated with T2DM pathogenesis, and to predict the underlying molecular mechanisms. As a result, a total of 1,512 differentially expressed (DE) lncRNAs and 1,923 DE mRNAs were identified through microarray analysis. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis indicated that multiple metabolic processes were dysregulated such as small molecule, organic acid, lipid and branched chain amino acid metabolism. Protein-protein interaction network was constructed and 10 hub mRNAs were identified, including EHHADH, ATM, ACOX1, PIK3R1, EGFR, UQCRFS1, HMGCL, UQCRC2, NDUFS3 and F2. RT-qPCR was conducted to verify the validity of microarray results. Then, coding-noncoding co-expression network and competing endogenous RNA (ceRNA) network were analyzed to predict the lncRNA-mRNA and lncRNA-miRNA-mRNA regulatory patterns. Subsequently, 10 key intermediating miRNAs in ceRNA networks with a node degree > 80 were identified, including hsa-miR-5692a, hsa-miR-12136, hsa-miR-5680, hsa-miR-1305, hsa-miR-6833-5p, hsa-miR-7159-5p, hsa-miR-548as-3p, hsa-miR-6873-3p, hsa-miR-1290 and hsa-miR-4768-5p. In conclusion, the present study evaluated the transcriptome profiling of lncRNAs and mRNAs in livers from T2DM patients, with a value for understanding the molecular mechanism of disease pathogenesis and identifying effective biomarkers in clinical diagnosis.
Collapse
Affiliation(s)
- Xi Lan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, grid.43169.39Xi'an Jiaotong University, Xi'an, China
| | - Jing Han
- Talent Highland and Center for Gut Microbiome Research of Med-X Institute, grid.452438.cFirst Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Binxian Wang
- Department of Microbiology and Immunology, School of Basic Medical Science, grid.43169.39Xi'an Jiaotong University, Xi'an, China
| | - Mingzhu Sun
- Department of Endocrinology, grid.452672.0Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
212
|
Chae HS, Dale O, Mir TM, Avula B, Zhao J, Khan IA, Khan SI. A Multitarget Approach to Evaluate the Efficacy of Aquilaria sinensis Flower Extract against Metabolic Syndrome. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27030629. [PMID: 35163893 PMCID: PMC8838142 DOI: 10.3390/molecules27030629] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/07/2022] [Accepted: 01/15/2022] [Indexed: 11/16/2022]
Abstract
Aquilaria sinensis (Lour.) Spreng is known for its resinous secretion (agarwood), often secreted in defense against injuries. We investigated the effects of A. sinensis flower extract (AF) on peroxisome proliferator-activated receptors alpha and gamma (PPARα and PPARγ), liver X receptor (LXR), glucose uptake, and lipid accumulation (adipogenesis). Activation of PPARα, PPARγ and LXR was determined in hepatic (HepG2) cells by reporter gene assays. Glucose uptake was determined in differentiated muscle (C2C12) cells using 2-NBDG (2-deoxy-2-[(7-nitro-2,1,3-benzoxadiazol-4-yl)amino]-D-glucose). Adipogenesis was determined in adipocytes (3T3-L1 cells) by Oil red O staining. At a concentration of 50 µg/mL, AF caused 12.2-fold activation of PPARα and 5.7-fold activation of PPARγ, while the activation of LXR was only 1.7-fold. AF inhibited (28%) the adipogenic effect induced by rosiglitazone in adipocytes and increased glucose uptake (32.8%) in muscle cells at 50 μg/mL. It was concluded that AF acted as a PPARα/γ dual agonist without the undesired effect of adipogenesis and exhibited the property of enhancing glucose uptake. This is the first report to reveal the PPARα/γ dual agonistic action and glucose uptake enhancing property of AF along with its antiadipogenic effect, indicating its potential in ameliorating the symptoms of metabolic syndrome.
Collapse
Affiliation(s)
- Hee-Sung Chae
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Oxford, MS 38677, USA; (H.-S.C.); (O.D.); (T.M.M.); (B.A.); (J.Z.); (I.A.K.)
| | - Olivia Dale
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Oxford, MS 38677, USA; (H.-S.C.); (O.D.); (T.M.M.); (B.A.); (J.Z.); (I.A.K.)
| | - Tahir Maqbool Mir
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Oxford, MS 38677, USA; (H.-S.C.); (O.D.); (T.M.M.); (B.A.); (J.Z.); (I.A.K.)
| | - Bharathi Avula
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Oxford, MS 38677, USA; (H.-S.C.); (O.D.); (T.M.M.); (B.A.); (J.Z.); (I.A.K.)
| | - Jianping Zhao
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Oxford, MS 38677, USA; (H.-S.C.); (O.D.); (T.M.M.); (B.A.); (J.Z.); (I.A.K.)
| | - Ikhlas A. Khan
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Oxford, MS 38677, USA; (H.-S.C.); (O.D.); (T.M.M.); (B.A.); (J.Z.); (I.A.K.)
- Department of Biomolecular Sciences, School of Pharmacy, The University of Mississippi, Oxford, MS 38677, USA
| | - Shabana I. Khan
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Oxford, MS 38677, USA; (H.-S.C.); (O.D.); (T.M.M.); (B.A.); (J.Z.); (I.A.K.)
- Department of Biomolecular Sciences, School of Pharmacy, The University of Mississippi, Oxford, MS 38677, USA
- Correspondence: ; Tel.: +1-662-915-1041
| |
Collapse
|
213
|
Fu Y, Yang K, Huang Y, Zhang Y, Li S, Li WD. Deciphering Risperidone-Induced Lipogenesis by Network Pharmacology and Molecular Validation. Front Psychiatry 2022; 13:870742. [PMID: 35509887 PMCID: PMC9058120 DOI: 10.3389/fpsyt.2022.870742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 03/18/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Risperidone is an atypical antipsychotic that can cause substantial weight gain. The pharmacological targets and molecular mechanisms related to risperidone-induced lipogenesis (RIL) remain to be elucidated. Therefore, network pharmacology and further experimental validation were undertaken to explore the action mechanisms of RIL. METHODS RILs were systematically analyzed by integrating multiple databases through integrated network pharmacology, transcriptomics, molecular docking, and molecular experiment analysis. The potential signaling pathways for RIL were identified and experimentally validated using gene ontology (GO) enrichment and Kyoto encyclopedia of genes and genomes (KEGG) analysis. RESULTS Risperidone promotes adipocyte differentiation and lipid accumulation through Oil Red O staining and reverse transcription-polymerase chain reaction (RT-PCR). After network pharmacology and GO analysis, risperidone was found to influence cellular metabolism. In addition, risperidone influences adipocyte metabolism, differentiation, and lipid accumulation-related functions through transcriptome analysis. Intersecting analysis, molecular docking, and pathway validation analysis showed that risperidone influences the adipocytokine signaling pathway by targeting MAPK14 (mitogen-activated protein kinase 14), MAPK8 (mitogen-activated protein kinase 8), and RXRA (retinoic acid receptor RXR-alpha), thereby inhibiting long-chain fatty acid β-oxidation by decreasing STAT3 (signal transducer and activator of transcription 3) expression and phosphorylation. CONCLUSION Risperidone increases adipocyte lipid accumulation by plausibly inhibiting long-chain fatty acid β-oxidation through targeting MAPK14 and MAPK8.
Collapse
Affiliation(s)
- Yun Fu
- Department of Genetics, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ke Yang
- Department of Genetics, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yepei Huang
- Department of Genetics, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yuan Zhang
- Department of Genetics, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Shen Li
- Department of Genetics, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.,Department of Psychiatry and Psychology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Wei-Dong Li
- Department of Genetics, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
214
|
Lin Y, Wang Y, Li PF. PPARα: An emerging target of metabolic syndrome, neurodegenerative and cardiovascular diseases. Front Endocrinol (Lausanne) 2022; 13:1074911. [PMID: 36589809 PMCID: PMC9800994 DOI: 10.3389/fendo.2022.1074911] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 11/23/2022] [Indexed: 12/23/2022] Open
Abstract
Peroxisome proliferator-activated receptor α (PPARα) is a ligand-activated transcription factor that is involved in lipid metabolism of various tissues. Different metabolites of fatty acids and agonists like fibrates activate PPARα for its transactivative or repressive function. PPARα is known to affect diverse human diseases, and we focus on advanced studies of its transcriptional regulation in these diseases. In MAFLD, PPARα shows a protective function with its upregulation of lipid oxidation and mitochondrial biogenesis and transcriptional repression of inflammatory genes, which is similar in Alzheimer's disease and cardiovascular disease. Activation of PPARα also prevents the progress of diabetes complications; however, its role in diabetes and cancers remains uncertain. Some PPARα-specific agonists, such as Wy14643 and fenofibrate, have been applied in metabolic syndrome treatment, which might own potential in wider application. Future studies may further explore the functions and interventions of PPARα in cancer, diabetes, immunological diseases, and neurodegenerative disease.
Collapse
Affiliation(s)
- Yijun Lin
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen, China
- *Correspondence: Yijun Lin, ; Yan Wang, ; Pei-feng Li,
| | - Yan Wang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen, China
- *Correspondence: Yijun Lin, ; Yan Wang, ; Pei-feng Li,
| | - Pei-feng Li
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen, China
- *Correspondence: Yijun Lin, ; Yan Wang, ; Pei-feng Li,
| |
Collapse
|
215
|
Rab2A regulates the progression of nonalcoholic fatty liver disease downstream of AMPK-TBC1D1 axis by stabilizing PPARγ. PLoS Biol 2022; 20:e3001522. [PMID: 35061665 PMCID: PMC8809606 DOI: 10.1371/journal.pbio.3001522] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 02/02/2022] [Accepted: 12/21/2021] [Indexed: 12/16/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) affects approximately a quarter of the population worldwide, and persistent overnutrition is one of the major causes. However, the underlying molecular basis has not been fully elucidated, and no specific drug has been approved for this disease. Here, we identify a regulatory mechanism that reveals a novel function of Rab2A in the progression of NAFLD based on energy status and PPARγ. The mechanistic analysis shows that nutrition repletion suppresses the phosphorylation of AMPK-TBC1D1 signaling, augments the level of GTP-bound Rab2A, and then increases the protein stability of PPARγ, which ultimately promotes the hepatic accumulation of lipids in vitro and in vivo. Furthermore, we found that blocking the AMPK-TBC1D1 pathway in TBC1D1S231A-knock-in (KI) mice led to a markedly increased GTP-bound Rab2A and subsequent fatty liver in aged mice. Our studies also showed that inhibition of Rab2A expression alleviated hepatic lipid deposition in western diet-induced obesity (DIO) mice by reducing the protein level of PPARγ and the expression of PPARγ target genes. Our findings not only reveal a new molecular mechanism regulating the progression of NAFLD during persistent overnutrition but also have potential implications for drug discovery to combat this disease. Non-alcoholic fatty liver disease (NAFLD) affects approximately a quarter of the global population; persistent overnutrition is one of the major causes, but the molecular mechanism remains unclear. This study shows that overnutrition suppresses the phosphorylation of AMPK and TBC1D1, augmenting the level of GTP-bound Rab2A and increasing the stability of PPARγ, which ultimately promotes the hepatic accumulation of lipids.
Collapse
|
216
|
Zhang M, Zhou Y, Xie Z, Luo S, Zhou Z, Huang J, Zhao B. New Developments in T Cell Immunometabolism and Therapeutic Implications for Type 1 Diabetes. Front Endocrinol (Lausanne) 2022; 13:914136. [PMID: 35757405 PMCID: PMC9226440 DOI: 10.3389/fendo.2022.914136] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/09/2022] [Indexed: 11/23/2022] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease mediated by T cells and is becoming a serious public health threat. Despite the increasing incidence rate of T1D worldwide, our understanding of why T1D develops and how T cells lose their self-tolerance in this process remain limited. Recent advances in immunometabolism have shown that cellular metabolism plays a fundamental role in shaping T cell responses. T cell activation and proliferation are supported by metabolic reprogramming to meet the increased energy and biomass demand, and deregulation in immune metabolism can lead to autoimmune disorders. Specific metabolic pathways and factors have been investigated to rectify known deficiencies in several autoimmune diseases, including T1D. Most therapeutic strategies have concentrated on aerobic glycolysis to limit T cell responses, whereas glycolysis is the main metabolic pathway for T cell activation and proliferation. The use of metabolic inhibitors, especially glycolysis inhibitors may largely leave T cell function intact but primarily target those autoreactive T cells with hyperactivated metabolism. In this review, we provide an overview of metabolic reprogramming used by T cells, summarize the recent findings of key metabolic pathways and regulators modulating T cell homeostasis, differentiation, and function in the context of T1D, and discuss the opportunities for metabolic intervention to be employed to suppress autoreactive T cells and limit the progression of β-cell destruction.
Collapse
Affiliation(s)
- Mengdi Zhang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yanyan Zhou
- Department of Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhiguo Xie
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shuoming Luo
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jiaqi Huang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Bin Zhao, ; ; Jiaqi Huang, ;
| | - Bin Zhao
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Bin Zhao, ; ; Jiaqi Huang, ;
| |
Collapse
|
217
|
Zhang YT, Zeeshan M, Su F, Qian ZM, Dee Geiger S, Edward McMillin S, Wang ZB, Dong PX, Ou YQ, Xiong SM, Shen XB, Zhou PE, Yang BY, Chu C, Li QQ, Zeng XW, Feng WR, Zhou YZ, Dong GH. Associations between both legacy and alternative per- and polyfluoroalkyl substances and glucose-homeostasis: The Isomers of C8 health project in China. ENVIRONMENT INTERNATIONAL 2022; 158:106913. [PMID: 34624590 DOI: 10.1016/j.envint.2021.106913] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 09/28/2021] [Accepted: 09/28/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Epidemiological studies on the associations of legacy per- and polyfluoroalkyl substances (PFASs) and glucose homeostasis remain discordant. Understanding of PFAS alternatives is limited, and few studies have reported joint associations of PFASs and PFAS alternatives. OBJECTIVES To investigate associations of novel PFAS alternatives (chlorinated perfluoroalkyl ether sulfonic acids, Cl-PFESAs and perfluorobutanoic acid, PFBA) and two legacy PFASs (Perfluorooctanoic acid, PFOA and perfluorooctane sulfonate, PFOS) with glucose-homeostasis markers and explore joint associations of 13 legacy and alternative PFASs with the selected outcomes. METHODS We used cross-sectional data of 1,038 adults from the Isomers of C8 Health Project in China. Associations of PFASs and PFAS alternatives with glucose-homeostasis were explored in single-pollutant models using generalized linear models with natural cubic splines for PFASs. Bayesian Kernel Machine Regression (BKMR) models were applied to assess joint associations of exposures and outcomes. Sex-specific analyses were also conducted to evaluate effect modification. RESULTS After adjusting for confounders, both legacy (PFOA, PFOS) and alternative (Cl-PFESAs and PFBA) PFASs were positively associated with glucose-homeostasis markers in single-pollutant models. For example, in the total study population, estimated changes with 95% confidence intervals (CI) of fasting glucose at the 95th percentile of 6:2Cl-PFESA and PFOS against the thresholds were 0.90 (95% CI: 0.59, 1.21) and 0.44 (95% CI: 0.26, 0.62). Positive joint associations were found in BKMR models with 6:2Cl-PFESA contributing most. Sex-specific associations existed in both single- and multi-pollutant models. CONCLUSIONS Legacy and alternative PFASs were positively associated with glucose-homeostasis markers. 6:2Cl-PFESA was the primary contributor. Sex-specific associations were also identified. These results indicate that joint associations and effect modification should be considered in risk assessment. However, further studies are recommended to strengthen our findings and to elucidate the mechanisms of action of legacy and alternative PFASs.
Collapse
Affiliation(s)
- Yun-Ting Zhang
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Mohammed Zeeshan
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Fan Su
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Zheng-Min Qian
- Department of Epidemiology and Biostatistics, College for Public Health & Social Justice, Saint Louis University, Saint Louis, MO 63104, USA
| | - Sarah Dee Geiger
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, IL 61820, USA
| | - Stephen Edward McMillin
- School of Social Work, College for Public Health and Social Justice, Saint Louis University, Saint Louis, MO 63103, USA
| | - Zhi-Bin Wang
- Department of Environmental Health Sciences, Laboratory of Human Environmental Epigenomes, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Peng-Xin Dong
- Nursing College, Guangxi Medical University, Nanning 530021, China
| | - Yan-Qiu Ou
- Department of Epidemiology, Guangdong Cardiovascular Institute, WHO Collaborating Center for Research and Training in Cardiovascular Diseases, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Shi-Min Xiong
- School of Public Health, Zunyi Medical University, Zunyi 563060, China
| | - Xu-Bo Shen
- School of Public Health, Zunyi Medical University, Zunyi 563060, China
| | - Pei-En Zhou
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Bo-Yi Yang
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Chu Chu
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Qing-Qing Li
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiao-Wen Zeng
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Wen-Ru Feng
- Department of Environmental Health, Guangzhou Center for Disease Control and Prevention, Guangzhou 510440, China.
| | - Yuan-Zhong Zhou
- School of Public Health, Zunyi Medical University, Zunyi 563060, China.
| | - Guang-Hui Dong
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
218
|
Rojas A, Lindner C, Schneider I, Gonzàlez I, Araya H, Morales E, Gómez M, Urdaneta N, Araya P, Morales MA. Diabetes mellitus contribution to the remodeling of the tumor microenvironment in gastric cancer. World J Gastrointest Oncol 2021; 13:1997-2012. [PMID: 35070037 PMCID: PMC8713306 DOI: 10.4251/wjgo.v13.i12.1997] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/10/2021] [Accepted: 10/27/2021] [Indexed: 02/06/2023] Open
Abstract
Compelling pieces of evidence derived from both clinical and experimental research has demonstrated the crucial contribution of diabetes mellitus (DM) as a risk factor associated with increased cancer incidence and mortality in many human neoplasms, including gastric cancer (GC). DM is considered a systemic inflammatory disease and therefore, this inflammatory status may have profound effects on the tumor microenvironment (TME), particularly by driving many molecular mechanisms to generate a more aggressive TME. DM is an active driver in the modification of the behavior of many cell components of the TME as well as altering the mechanical properties of the extracellular matrix (ECM), leading to an increased ECM stiffening. Additionally, DM can alter many cellular signaling mechanisms and thus favoring tumor growth, invasion, and metastatic potential, as well as key elements in regulating cellular functions and cross-talks, such as the microRNAs network, the production, and cargo of exosomes, the metabolism of cell stroma and resistance to hypoxia. In the present review, we intend to highlight the mechanistic contributions of DM to the remodeling of TME in GC.
Collapse
Affiliation(s)
- Armando Rojas
- Biomedical Research Lab., Medicine Faculty, Catholic University of Maule, Talca 34600000, Chile
| | - Cristian Lindner
- Biomedical Research Lab., Medicine Faculty, Catholic University of Maule, Talca 34600000, Chile
| | - Iván Schneider
- Biomedical Research Lab., Medicine Faculty, Catholic University of Maule, Talca 34600000, Chile
| | - Ileana Gonzàlez
- Biomedical Research Lab., Medicine Faculty, Catholic University of Maule, Talca 34600000, Chile
| | - Hernan Araya
- Department of Clinical Sciences, Medicine Faculty, Catholic University of Maule, Talca 34600000, Chile
- Servicio de Oncología, Hospital Regional de Talca, Talca 34600000, Chile
| | - Erik Morales
- Biomedical Research Lab., Medicine Faculty, Catholic University of Maule, Talca 34600000, Chile
- Servicio de Anatomía Patologica, Hospital Regional de Talca, Talca 34600000, Chile
| | - Milibeth Gómez
- Department of Clinical Sciences, Medicine Faculty, Catholic University of Maule, Talca 34600000, Chile
- Servicio de Oncología, Hospital Regional de Talca, Talca 34600000, Chile
| | - Nelson Urdaneta
- Department of Clinical Sciences, Medicine Faculty, Catholic University of Maule, Talca 34600000, Chile
- Servicio de Oncología, Hospital Regional de Talca, Talca 34600000, Chile
| | - Paulina Araya
- Biomedical Research Lab., Medicine Faculty, Catholic University of Maule, Talca 34600000, Chile
| | - Miguel Angel Morales
- Department of Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, University of Chile, Santiago 8320000, Chile
| |
Collapse
|
219
|
Vales-Villamarín C, de Dios O, Pérez-Nadador I, Gavela-Pérez T, Soriano-Guillén L, Garcés C. PPARγ2 Pro12Ala Polymorphism is Associated in Children With Traits Related to Susceptibility to Type 2 Diabetes. Front Pharmacol 2021; 12:763853. [PMID: 34887761 PMCID: PMC8650059 DOI: 10.3389/fphar.2021.763853] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 11/10/2021] [Indexed: 12/16/2022] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is a ligand-activated nuclear receptor that regulates glucose and lipid metabolism. Pharmacological activators of PPARγ are being used as a treatment of obesity related disorders such as dyslipidaemia and type 2 diabetes, but questions remain open regarding the effects of PPARγ on traits related to the development of type 2 diabetes. In our study, we have analyzed the relationship of the common variant Pro12Ala in the human PPARγ2 gene with the presence of obesity and with insulin, HOMA and lipid profile in a representative sample of 6-to 8-year-old children free from the confounding factors associated with adults. We found that Ala12Ala genotype was significantly more frequent in females with obesity than in those without obesity, with Ala12Ala carriers having significantly higher weight and body mass index (BMI), however the association disappeared when adjusting by leptin concentrations. The Ala12Ala genotype was associated with significantly higher HDL-cholesterol and apoA-I levels in males but not in females, independently of BMI. In a recessive model, in females, leptin levels appeared higher in Ala12Ala carriers. Although no apparent differences were observed in any sex when analyzing insulin levels and HOMA among genotypes without adjusting, lower insulin levels and lower HOMA appeared associated with Ala12Ala carriers when adjusting for BMI and leptin levels. In summary, our data showed that leptin seems to be having an effect on the association between the PPARγ2 Pro12Ala and BMI. Besides, after controlling for BMI and leptin, a protective effect of the Ala12Ala variant of the PPARγ2 Pro12Ala polymorphism on insulin sensitivity is evident already in prepubertal children.
Collapse
Affiliation(s)
| | - Olaya de Dios
- Lipid Research Laboratory, IIS-Fundación Jiménez Díaz UAM, Madrid, Spain
| | - Iris Pérez-Nadador
- Lipid Research Laboratory, IIS-Fundación Jiménez Díaz UAM, Madrid, Spain
| | | | | | - Carmen Garcés
- Lipid Research Laboratory, IIS-Fundación Jiménez Díaz UAM, Madrid, Spain
| |
Collapse
|
220
|
Li Z, Ren Q, Zhou Z, Cai Z, Wang B, Han J, Zhang L. Discovery of the first-in-class dual PPARδ/γ partial agonist for the treatment of metabolic syndrome. Eur J Med Chem 2021; 225:113807. [PMID: 34455359 DOI: 10.1016/j.ejmech.2021.113807] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/24/2021] [Accepted: 08/24/2021] [Indexed: 12/29/2022]
Abstract
The peroxisome proliferator-activated receptors (PPARs) exert vital function in the regulation of energy metabolism, which were considered as promising targets of metabolic syndrome. Until now, PPARδ/γ dual agonist is rarely reported, and thereby the pharmacologic action of PPARδ/γ dual agonist is still unclear. In this study, we identified a dual PPARδ/γ partial agonist 6 (ZLY06) based on the cyclization strategy of PPARα/δ dual agonist GFT505. ZLY06 revealed excellent pharmacokinetic profiles suitable for oral medication. Moreover, ZLY06 markedly improved glucolipid metabolism without weight gain, and alleviated fatty liver by promoting the β-oxidation of fatty acid and inhibiting hepatic lipogenesis. In contrast, weight gain and hepatic steatosis were observed in Rosiglitazone, a widely used PPARγ full agonist. All of these results indicated that ZLY06 exhibits potential benefits on metabolic syndrome, while no adverse effects related to PPARγ full agonist.
Collapse
Affiliation(s)
- Zheng Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of New Drug Discovery and Evaluation of Ordinary Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Qiang Ren
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of New Drug Discovery and Evaluation of Ordinary Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Zongtao Zhou
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Zongyu Cai
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Bin Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Jing Han
- School of Chemistry and Chemical Engineering, Jiangsu Key Laboratory of Green Synthetic Chemistry for Functional Materials, Jiangsu Normal University, Xuzhou, 221116, PR China
| | - Luyong Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of New Drug Discovery and Evaluation of Ordinary Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, 210009, PR China.
| |
Collapse
|
221
|
Niu L, Hou Y, Jiang M, Bai G. The rich pharmacological activities of Magnolia officinalis and secondary effects based on significant intestinal contributions. JOURNAL OF ETHNOPHARMACOLOGY 2021; 281:114524. [PMID: 34400262 DOI: 10.1016/j.jep.2021.114524] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 08/01/2021] [Accepted: 08/13/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Magnolia officinalis Cortex (M. officinalis) is a traditional herbal drug widely used in Asian countries. Depending on its multiple biological activities, M. officinalis is used to regulate gastrointestinal (GI) motility, relieve cough and asthma, prevent cardiovascular and cerebrovascular diseases, and treat depression and anxiety. AIM OF THE REVIEW We aimed to review the abundant form of pharmacodynamics activity and potential mechanisms of action of M. officinalis and the characteristics of the internal processes of the main components. The potential mechanisms of local and distance actions of M. officinalis based on GI tract was provided, and it was used to reveal the interconnections between traditional use, phytochemistry, and pharmacology. MATERIALS AND METHODS Published literatures about M. officinalis and its main components were collected from several scientific databases, including PubMed, Elsevier, ScienceDirect, Google Scholar and Web of Science etc. RESULTS: M. officinalis was shown multiple effects including effects on digestive system, respiratory system, central system, which is consistent with traditional applications, as well as some other activities such as cardiovascular system, anticancer, anti-inflammatory and antioxidant effects and so on. The mechanisms of these activities are abundant. Its chief ingredients such as magnolol and honokiol can be metabolized into active metabolites in vivo, which can increase water solubility and bioavailability and exert pharmacological activity in the whole body. In the GI tract, M. officinalis and its main ingredient can regulate GI hormones and substance metabolism, protect the intestinal barrier and affect the gut microbiota (GM). These actions are effective to improve local discomfort and some distal symptoms such as depression, asthma, or metabolic disorders. CONCLUSIONS Although M. officinalis has rich pharmacological effects, the GI tract makes great contributions to it. The GI tract is not only an important place for absorption and metabolism but also a key site to help M. officinalis exert local and distal efficacy. Pharmacodynamical studies on the efficacies of distal tissues based on the contributions of the GI tract hold great potential for understanding the benefits of M. officinalis and providing new ideas for the treatment of important diseases.
Collapse
Affiliation(s)
- Lin Niu
- Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, People's Republic of China
| | - Yuanyuan Hou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, People's Republic of China
| | - Min Jiang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, People's Republic of China
| | - Gang Bai
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, People's Republic of China.
| |
Collapse
|
222
|
Montaigne D, Butruille L, Staels B. PPAR control of metabolism and cardiovascular functions. Nat Rev Cardiol 2021; 18:809-823. [PMID: 34127848 DOI: 10.1038/s41569-021-00569-6] [Citation(s) in RCA: 359] [Impact Index Per Article: 119.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/10/2021] [Indexed: 12/22/2022]
Abstract
Peroxisome proliferator-activated receptor-α (PPARα), PPARδ and PPARγ are transcription factors that regulate gene expression following ligand activation. PPARα increases cellular fatty acid uptake, esterification and trafficking, and regulates lipoprotein metabolism genes. PPARδ stimulates lipid and glucose utilization by increasing mitochondrial function and fatty acid desaturation pathways. By contrast, PPARγ promotes fatty acid uptake, triglyceride formation and storage in lipid droplets, thereby increasing insulin sensitivity and glucose metabolism. PPARs also exert antiatherogenic and anti-inflammatory effects on the vascular wall and immune cells. Clinically, PPARγ activation by glitazones and PPARα activation by fibrates reduce insulin resistance and dyslipidaemia, respectively. PPARs are also physiological master switches in the heart, steering cardiac energy metabolism in cardiomyocytes, thereby affecting pathological heart failure and diabetic cardiomyopathy. Novel PPAR agonists in clinical development are providing new opportunities in the management of metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- David Montaigne
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Laura Butruille
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Bart Staels
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France.
| |
Collapse
|
223
|
Zhu J, Yu C, Zhou H, Wei X, Wang Y. Comparative evaluation for phytochemical composition and regulation of blood glucose, hepatic oxidative stress and insulin resistance in mice and HepG2 models of four typical Chinese dark teas. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2021; 101:6563-6577. [PMID: 34018615 DOI: 10.1002/jsfa.11328] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 04/13/2021] [Accepted: 05/21/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Dark tea, comprising one of the six major teas, has many biological activities, which originate from their active substrates, such as polyphenols, polysaccharides, and so on. The hypoglycemic effect is one of its most prominent activities, although less is known about their evaluation and potential role in the hypoglycemic mechanism. RESULTS In the present study, we separately analyzed the phytochemical composition, glycosidase inhibition and free radical scavenging activities, and hypoglycemic activity in type 2 diabetes mellitus mice, as well as the alleviation of insulin resistance in HepG2 cells of four dark tea aqueous extracts. The results showed that the phytochemical composition of dark tea aqueous extracts was significantly different, and they all had good glycosidase inhibition and free radical scavenging activities, in vivo hypoglycemic activity and alleviation of insulin resistance, and could also activate the phosphatidylinositol 3-kinase-Akt-perixisome proliferation-activated receptor cascade signaling pathway to regulate glucose and lipid metabolism, change the key enzyme activities related to glucose metabolism and antioxidant activity, and reduce oxidative stress and inflammatory factor levels. Among them, Liubao brick tea (LBT) and Pu-erh tea (PET) possessed better glycosidase inhibitory activity, in vivo hypoglycemic activity and improved insulin resistance activity, whereas Qingzhuan brick tea and Fuzhuan brick tea had better free radical scavenging activity, which may be explained by their distinct phytochemical compositions, such as tea proteins, polysaccharides, polyphenols, catechins, and tea pigments and some elements. CONCLUSION Dark tea is a highly attractive candidate for developing antidiabetic food, LBT and PET may be good natural sources of agricultural products with anti-diabetic effects. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Jiangxiong Zhu
- Institute of Food Engineering, College of Life Science, Shanghai Normal University, Shanghai, China
| | - Chuang Yu
- Institute of Food Engineering, College of Life Science, Shanghai Normal University, Shanghai, China
| | - Hui Zhou
- Institute of Food Engineering, College of Life Science, Shanghai Normal University, Shanghai, China
| | - Xinlin Wei
- Institute of Food Engineering, College of Life Science, Shanghai Normal University, Shanghai, China
- Department of Food Science & Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Yuanfeng Wang
- Institute of Food Engineering, College of Life Science, Shanghai Normal University, Shanghai, China
| |
Collapse
|
224
|
Dixit G, Prabhu A. The pleiotropic peroxisome proliferator activated receptors: Regulation and therapeutics. Exp Mol Pathol 2021; 124:104723. [PMID: 34822814 DOI: 10.1016/j.yexmp.2021.104723] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/02/2021] [Accepted: 11/15/2021] [Indexed: 02/07/2023]
Abstract
The Peroxisome proliferator-activated receptors (PPARs) are key regulators of metabolic events in our body. Owing to their implication in maintenance of homeostasis, both PPAR agonists and antagonists assume therapeutic significance. Understanding the molecular mechanisms of each of the PPAR isotypes in the healthy body and during disease is crucial to exploiting their full therapeutic potential. This article is an attempt to present a rational analysis of the multifaceted therapeutic effects and underlying mechanisms of isotype-specific PPAR agonists, dual PPAR agonists, pan PPAR agonists as well as PPAR antagonists. A holistic understanding of the mechanistic dimensions of these key metabolic regulators will guide future efforts to identify novel molecules in the realm of metabolic, inflammatory and immunotherapeutic diseases.
Collapse
Affiliation(s)
- Gargi Dixit
- Department of Pharmaceutical Chemistry & Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Arati Prabhu
- Department of Pharmaceutical Chemistry & Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India.
| |
Collapse
|
225
|
Oh KK, Adnan M, Cho DH. Elucidating Drug-Like Compounds and Potential Mechanisms of Corn Silk ( Stigma Maydis) against Obesity: A Network Pharmacology Study. Curr Issues Mol Biol 2021; 43:1906-1936. [PMID: 34889899 PMCID: PMC8929052 DOI: 10.3390/cimb43030133] [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: 09/23/2021] [Revised: 10/27/2021] [Accepted: 11/03/2021] [Indexed: 11/16/2022] Open
Abstract
Corn silk (Stigma Maydis) has been utilized as an important herb against obesity by Chinese, Korean, and Native Americans, but its phytochemicals and mechanisms(s) against obesity have not been deciphered completely. This study aimed to identify promising bioactive constituents and mechanism of action(s) of corn silk (CS) against obesity via network pharmacology. The compounds from CS were identified using Gas Chromatography Mass Spectrometry (GC-MS) and were confirmed ultimately by Lipinski's rule via SwissADME. The relationships of the compound-targets or obesity-related targets were confirmed by public bioinformatics. The signaling pathways related to obesity, protein-protein interaction (PPI), and signaling pathways-targets-bioactives (STB) were constructed, visualized, and analyzed by RPackage. Lastly, Molecular Docking Test (MDT) was performed to validate affinity between ligand(s) and protein(s) on key signaling pathway(s). We identified a total of 36 compounds from CS via GC-MS, all accepted by Lipinski's rule. The number of 36 compounds linked to 154 targets, 85 among 154 targets related directly to obesity-targets (3028 targets). Of the final 85 targets, we showed that the PPI network (79 edges, 357 edges), 12 signaling pathways on a bubble chart, and STB network (67 edges, 239 edges) are considered as therapeutic components. The MDT confirmed that two key activators (β-Amyrone, β-Stigmasterol) bound most stably to PPARA, PPARD, PPARG, FABP3, FABP4, and NR1H3 on the PPAR signaling pathway, also, three key inhibitors (Neotocopherol, Xanthosine, and β-Amyrone) bound most tightly to AKT1, IL6, FGF2, and PHLPP1 on the PI3K-Akt signaling pathway. Overall, we provided promising key signaling pathways, targets, and bioactives of CS against obesity, suggesting crucial pharmacological evidence for further clinical testing.
Collapse
Affiliation(s)
| | | | - Dong-Ha Cho
- Department of Bio-Health Convergence, College of Biomedical Science, Kangwon National University, Chuncheon 24341, Korea; (K.-K.O.); (M.A.)
| |
Collapse
|
226
|
Chen M, Jing D, Ye R, Yi J, Zhao Z. PPARβ/δ accelerates bone regeneration in diabetic mellitus by enhancing AMPK/mTOR pathway-mediated autophagy. Stem Cell Res Ther 2021; 12:566. [PMID: 34736532 PMCID: PMC8567548 DOI: 10.1186/s13287-021-02628-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 10/16/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Diabetic patients are more vulnerable to skeletal complications. Peroxisome proliferators-activated receptor (PPAR) β/δ has a positive regulatory effect on bone turnover under physiologic glucose concentration; however, the regulatory effect in diabetes mellitus has not been investigated yet. Herein, we explored the effects of PPARβ/δ agonist on the regeneration of diabetic bone defects and the osteogenic differentiation of rat bone marrow mesenchymal stem cells (rBMSCs) under a pathological high-glucose condition. METHODS We detected the effect of PPARβ/δ agonist on osteogenic differentiation of rBMSCs in vitro and investigated the bone healing process in diabetic rats after PPARβ/δ agonist treatment in vivo. RNA sequencing was performed to detect the differentially expressed genes and enriched pathways. Western blot was performed to detect the autophagy-related protein level. Laser confocal microscope (LSCM) and transmission electron microscope (TEM) were used to observe the formation of autophagosomes. RESULTS Our results demonstrated that the activation of PPARβ/δ can improve the osteogenic differentiation of rBMSCs in high-glucose condition and promote the bone regeneration of calvarial defects in diabetic rats, while the inhibition of PPARβ/δ alleviated the osteogenic differentiation of rBMSCs. Mechanistically, the activation of PPARβ/δ up-regulates AMPK phosphorylation, yielding mTOR suppression and resulting in enhanced autophagy activity, which further promotes the osteogenic differentiation of rBMSCs in high-glucose condition. The addition of AMPK inhibitor Compound C or autophagy inhibitor 3-MA inhibited the osteogenesis of rBMSCs in high-glucose condition, suggesting that PPARβ/δ agonist promotes osteogenic differentiation of rBMSCs through AMPK/mTOR-regulated autophagy. CONCLUSION In conclusion, our study demonstrates the potential role of PPARβ/δ as a molecular target for the treatment of impaired bone quality and delayed bone healing in diabetic patients for the first time.
Collapse
Affiliation(s)
- Miao Chen
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, No. 14, 3rd Section, South Renmin Road, Chengdu, 610041, Sichuan, China
| | - Dian Jing
- Department of Orthodontics, Shanghai Ninth People's Hospital, Collage of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Ye
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, No. 14, 3rd Section, South Renmin Road, Chengdu, 610041, Sichuan, China
| | - Jianru Yi
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China. .,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, No. 14, 3rd Section, South Renmin Road, Chengdu, 610041, Sichuan, China.
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China. .,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, No. 14, 3rd Section, South Renmin Road, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
227
|
Zhai Z, Niu KM, Liu H, Lin C, Tu Y, Liu Y, Cai L, Ouyang K, Liu J. Policosanol alleviates hepatic lipid accumulation by regulating bile acids metabolism in C57BL6/mice through AMPK-FXR-TGR5 cross-talk. J Food Sci 2021; 86:5466-5478. [PMID: 34730235 DOI: 10.1111/1750-3841.15951] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 09/13/2021] [Accepted: 09/24/2021] [Indexed: 12/22/2022]
Abstract
Policosanol exhibits a lipid accumulation alleviating effect, but the underlying mechanisms remains unclear. Bile acids are a significant factor in regulating cholesterol and lipid metabolism homeostasis in mammals. This study was aimed to elucidate the alleviating effect and underlying mechanisms of policosanol on hepatic lipid accumulation through bile acid (BA) metabolism. Policosanol supplementation significantly reduced hepatic triglycerides (19.29%), cholesterol (30.38%) in high fat diet (HFD) induced obese mice (P < 0.05). Furthermore, compared with the control group, HFD decreased the levels of total BAs (TBAs, 37.67%) and cholic acid (CA, 62.74%) in the serum of mice (P < 0.05). Meanwhile, compared to HFD group, policosanol also increased the level of secondary BAs (SBAs) and muricholic acids (MCAs, P < 0.05). qRT-PCR combined with protein level analysis revealed that policosanol significantly decreased sterol regulatory element-binding protein (SREBP-1c) and CD36, and increased the expression level of cytochrome P450 family 7 subfamily A member 1 (CYP7A1) and cytochrome P450 Family 27 Subfamily A Member 1 (CYP27A1, P < 0.05). Additionally, in the liver, policosanol was found downregulated the expression of farnesoid X receptor (FXR)-small heterodimer partner (SHP), and activate the Takeda G-coupled protein receptor 5 (TGR5)-adenosine-monophosphate-activated protein kinase (APMK) signaling pathway (P < 0.05). Peroxisome proliferator activated receptor (PPAR)-α, hormone sensitive lipase (HSL), and carnitine palmitoyltransferase (CPT)-1α also significantly increased in HP group (P < 0.05). The aforementioned results reveal that the potential mechanism of policosanol in alleviating liver lipid accumulation is to promote BA synthesis and lipolysis through regulating the cross-talk of the AMPK-FXR-TGR5. New insight for the application of policosanol as an anti-fatty liver functional food ingredient or supplement is also provided. PRACTICAL APPLICATION: Policosanol is an important active component of cereals and insect waxes (15-80%). However, almost no policosanol in refined foods such as clear corn germ oil and wheat flour. This study showed that oral administration of policosanol can significantly reduce triglyceride and cholesterol levels in the liver through affecting AMPK-TGR5-FXR cross-talk, whereas no significant toxicological effect is reported in human and mouse models. This study may provide theoretical support for the theory of dietary structure and the development of dietary supplements to improve lipid metabolism targeting the "bile acid-AMPK-TGR5" pathway.
Collapse
Affiliation(s)
- Zhenya Zhai
- Jiangxi Functional Feed Additive Engineering Laboratory, Institute of Biological Resource, Jiangxi Academy of Sciences, Nanchang, China.,CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, China
| | - Kai-Min Niu
- Jiangxi Functional Feed Additive Engineering Laboratory, Institute of Biological Resource, Jiangxi Academy of Sciences, Nanchang, China.,CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, China
| | - Huiping Liu
- Era Biotechnology (Shenzhen) Co., Ltd., Shenzhen, China
| | - Chong Lin
- Jiangxi Functional Feed Additive Engineering Laboratory, Institute of Biological Resource, Jiangxi Academy of Sciences, Nanchang, China
| | - Yue Tu
- Jiangxi Functional Feed Additive Engineering Laboratory, Institute of Biological Resource, Jiangxi Academy of Sciences, Nanchang, China
| | - Yichun Liu
- Jiangxi Functional Feed Additive Engineering Laboratory, Institute of Biological Resource, Jiangxi Academy of Sciences, Nanchang, China
| | - Lichuang Cai
- Jiangxi Functional Feed Additive Engineering Laboratory, Institute of Biological Resource, Jiangxi Academy of Sciences, Nanchang, China
| | - Kexian Ouyang
- Jiangxi Functional Feed Additive Engineering Laboratory, Institute of Biological Resource, Jiangxi Academy of Sciences, Nanchang, China
| | - Jianping Liu
- Jiangxi Functional Feed Additive Engineering Laboratory, Institute of Biological Resource, Jiangxi Academy of Sciences, Nanchang, China
| |
Collapse
|
228
|
Farhat F, Rahmi E, Chrestella J, Williamson O, Syari RP. Expressions of Nuclear Factor-kappa B and Peroxisome Proliferator-activated Receptor-Gamma Proportional with Clinical Staging of Nasopharyngeal Carcinoma. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.6261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: Nasopharyngeal carcinoma (NPC) is a malignancy induced by the mutation of the transcription factors nuclear factor-kappa B (NF-kB) and peroxisome proliferator-activated receptor-gamma (PPAR-gamma). There was no known of the study about the association and targeted therapy of NF-kB and PPAR-gamma-induced NPC.
AIM: This study analyzed and compared the proportion of NF-kB and PPAR-gamma and its association with the clinical characteristic of various NPC patients.
METHODS: This was a cross-sectional study and conducted in Adam Malik General Hospital. The samples were paraffin block tissue obtained from 58 NPC patients and underwent immunohistochemistry staining for NF-kB or PPAR-gamma overexpression in March–November 2018. Determination of overexpression was based on the immunoreactive score. The association of NF-kB or PPAR-gamma overexpression with the clinical characteristics of the patients was analyzed using Fisher’s exact test.
RESULTS: This study showed a significant increase of NF-kB and PPAR-gamma (p < 0.05). Male was found common than women (3.46:1) with non-keratinizing squamous cell carcinoma as the most common form of NPC (75.9%) and the 41–60 years old is the most common age (56.9%). Overexpression of NF-kB and PPAR-gamma was found mostly in T3-T4 (66.0%; 69.6%), N+ (92.5%; 91.3%), and clinical Stage IV (67.9%; 73.9%), respectively.
CONCLUSION: The number of samples overexpressed was proportional to the clinical stage of NPC. This study provides an insight into the relationship of NF-kB and PPAR-gamma to NPC, suggesting their role in the development of malignancy.
Collapse
|
229
|
Li X, Ge J, Li Y, Cai Y, Zheng Q, Huang N, Gu Y, Han Q, Li Y, Sun R, Liu R. Integrative lipidomic and transcriptomic study unravels the therapeutic effects of saikosaponins A and D on non-alcoholic fatty liver disease. Acta Pharm Sin B 2021; 11:3527-3541. [PMID: 34900534 PMCID: PMC8642447 DOI: 10.1016/j.apsb.2021.03.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/03/2021] [Accepted: 02/09/2021] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) has become one of the most prominent causes of chronic liver diseases and malignancies. However, few therapy has been approved. Radix Bupleuri (RB) is the most frequently used herbal medicine for the treatment of liver diseases. In the current study, we aim to systemically evaluate the therapeutic effects of saikosaponin A (SSa) and saikosaponin D (SSd), the major bioactive monomers in RB, against NAFLD and to investigate the underlying mechanisms. Our results demonstrated that both SSa and SSd improved diet-induced NAFLD. Integrative lipidomic and transcriptomic analysis revealed that SSa and SSd modulated glycerolipid metabolism by regulating related genes, like Lipe and Lipg. SSd profoundly suppressed the fatty acid biosynthesis by downregulating Fasn and Acaca expression and promoted fatty acid degradation by inducing Acox1 and Cpt1a expression. Bioinformatic analysis further predicted the implication of master transcription factors, including peroxisome proliferator-activated receptor alpha (PPARα), in the protective effects of SSa and SSd. These results were further confirmed in vitro in mouse primary hepatocytes. In summary, our study uncoded the complicated mechanisms underlying the promising anti-steatosis activities of saikosaponins (SSs), and provided critical evidence inspiring the discovery of innovative therapies based on SSa and SSd for the treatment of NAFLD and related complications.
Collapse
|
230
|
Steinman JB, Salomao MA, Pajvani UB. Zonation in NASH - A key paradigm for understanding pathophysiology and clinical outcomes. Liver Int 2021; 41:2534-2546. [PMID: 34328687 DOI: 10.1111/liv.15025] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 12/11/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) exists as a spectrum ranging from simple steatosis to histologically defined hepatocyte injury and inflammatory changes that define steatohepatitis (NASH), and increase risk for fibrosis. Although zonal differences in NASH have not been systematically studied, periportal involvement has been associated with worse metabolic outcomes and more hepatic fibrosis as compared to pericentral disease. These data suggest that hepatic zonation of disease may influence the diversity of clinical presentations. Similarly, several randomized clinical trials suggest a differential response based on zonation of disease, with preferential effects on periportal (cysteamine) or pericentral disease (obeticholic acid, pioglitazone). Intriguingly, morphogenic pathways known to affect zonal development and maintenance - WNT/β-Catenin, Hedgehog, HIPPO/Yap/TAZ and Notch - have been implicated in NASH pathogenesis, and nuclear hormone receptors downstream of potential NASH therapeutics show zonal preferences. In this review, we summarize these data and propose that patient-specific activation of these pathways may explain the variability in clinical presentation, and the zone-specific response observed in clinical trials.
Collapse
Affiliation(s)
| | - Marcela A Salomao
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, AZ, USA
| | - Utpal B Pajvani
- Department of Medicine, Columbia University, New York, NY, USA
| |
Collapse
|
231
|
Salomão R, Neto IVDS, Ramos GV, Tibana RA, Durigan JQ, Pereira GB, Franco OL, Royer C, Neves FDAR, de Carvalho ACA, Nóbrega OT, Haddad R, Prestes J, Marqueti RDC. Paternal Resistance Exercise Modulates Skeletal Muscle Remodeling Pathways in Fathers and Male Offspring Submitted to a High-Fat Diet. Front Physiol 2021; 12:706128. [PMID: 34646148 PMCID: PMC8503191 DOI: 10.3389/fphys.2021.706128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 09/02/2021] [Indexed: 11/24/2022] Open
Abstract
Although some studies have shown that a high-fat diet (HFD) adversely affects muscle extracellular matrix remodeling, the mechanisms involved in muscle trophism, inflammation, and adipogenesis have not been fully investigated. Thus, we investigated the effects of 8 weeks of paternal resistance training (RT) on gene and protein expression/activity of critical factors involved in muscle inflammation and remodeling of fathers and offspring (offspring exposed to standard chow or HFD). Animals were randomly distributed to constitute sedentary fathers (SF; n = 7; did not perform RT) or trained fathers (TF n = 7; performed RT), with offspring from mating with sedentary females. After birth, 28 male pups were divided into four groups (n = 7 per group): offspring from sedentary father submitted either to control diet (SFO-C) or high-fat diet (SFO-HF) and offspring from trained father submitted to control diet (TFO-C) or high-fat diet (TFO-HF). Our results show that an HFD downregulated collagen mRNA levels and upregulated inflammatory and atrophy pathways and adipogenic transcription factor mRNA levels in offspring gastrocnemius muscle. In contrast, paternal RT increased MMP-2 activity and decreased IL-6 levels in offspring exposed to a control diet. Paternal RT upregulated P70s6k and Ppara mRNA levels and downregulated Atrogin1 mRNA levels, while decreasing NFκ-B, IL-1β, and IL-8 protein levels in offspring exposed to an HFD. Paternal physical training influences key skeletal muscle remodeling pathways and inflammatory profiles relevant for muscle homeostasis maintenance in offspring submitted to different diets.
Collapse
Affiliation(s)
- Rebecca Salomão
- Laboratory of Molecular Analysis, Faculty of Ceilândia, Universidade de Brasília, Brasília, Brazil.,Graduate Program in Rehabilitation Sciences, Universidade de Brasília, Brasília, Brazil
| | - Ivo Vieira de Sousa Neto
- Laboratory of Molecular Analysis, Faculty of Ceilândia, Universidade de Brasília, Brasília, Brazil.,Graduate Program of Sciences and Technology of Health, Universidade de Brasília, Brasília, Brazil
| | | | - Ramires Alsamir Tibana
- Graduate Program in Health Sciences, Faculdade de Medicine, Universidade Federal do Mato Grosso (UFTM), Cuiabá, Brazil
| | | | - Guilherme Borges Pereira
- Interinstitutional Program of Post-Graduation in Physiological Sciences (UFSCar/UNESP), Department of Physiological Sciences, Universidade Federal de São Carlos, São Carlos, Brazil
| | - Octávio Luiz Franco
- Graduate Program in Genomics Science and Biotechnology, Universidade Católica de Brasília, Brasília, Brazil.,S-Inova Biotech, Graduate Program in Biotechnology, Universidade Católica Dom Bosco, Campo Grande, Brazil
| | - Carine Royer
- Laboratory of Molecular Analysis, Faculty of Ceilândia, Universidade de Brasília, Brasília, Brazil.,Laboratory of Molecular Pharmacology, Faculty of Health Sciences, Universidade de Brasília, Brasília, Brazil
| | | | | | - Otávio Toledo Nóbrega
- Graduate Program of Medical Sciences, Universidade de Brasília, Brasília, Brazil.,Center for Tropical Medicine, Universidade de Brasília, Brasília, Brazil
| | - Rodrigo Haddad
- Laboratory of Molecular Analysis, Faculty of Ceilândia, Universidade de Brasília, Brasília, Brazil.,Center for Tropical Medicine, Universidade de Brasília, Brasília, Brazil
| | - Jonato Prestes
- Graduate Program of Physical Education, Universidade Católica de Brasilia, Brasília, Brazil
| | - Rita de Cássia Marqueti
- Laboratory of Molecular Analysis, Faculty of Ceilândia, Universidade de Brasília, Brasília, Brazil.,Graduate Program in Rehabilitation Sciences, Universidade de Brasília, Brasília, Brazil.,Graduate Program of Sciences and Technology of Health, Universidade de Brasília, Brasília, Brazil
| |
Collapse
|
232
|
Chen K, Qu J, Chen H, Wang J, Hua H, Li J, Zhou L, Zhang W, Li Z. Investigating the medicinal potential, material basis and mechanism of Polygoni Orientalis Fructus based on multi-technology integrated network pharmacology. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 91:153685. [PMID: 34339945 DOI: 10.1016/j.phymed.2021.153685] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/20/2021] [Accepted: 07/17/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Polygoni Orientalis Fructus (POF) refers to the dried ripe fruit of Polygonum orientale L. which has a long historical application in clinic for treatment of various conditions in China. However, its chemical constituents, pharmacological effects and their coupled correlation have not been intensively investigated. PURPOSE In present work, we aimed to elucidate the medicinal material basis, optimum indication and corresponding therapeutic mechanism of POF. METHODS The main phytochemical ingredients in POF were characterized by liquid chromatography-mass spectrometry (LC-MS) analysis. The optimum medicinal potential and corresponding molecular mechanism of POF were deduced based on integrated statistic pattern recognition and network pharmacology. The deduced pharmacologic efficacy and mechanism of POF were further validated through in vitro study in free-fatty acid (FFA)-induced LO2 cells. RESULTS Total 30 main phytochemical ingredients were identified in POF in which 18 ingredients were screened to yield 277 potential targets. Based on analyzing the quantitative data matrix of drug-disease targets by statistic pattern recognition, non-alcoholic fatty liver disease (NAFLD) was screened as the optimum indication of POF from 23 candidate diseases. Promising action targets (PPARG, IL6, TNF, IL1B, IKBKB, RELA, etc.) and signaling pathways (AMPK signaling pathway, NF-κB signaling pathway, etc.) were screened and refined to elucidate the therapeutic mechanism of POF against NAFLD based on network pharmacology. In vitro study demonstrated that POF effectively alleviated FFA-induced steatosis, oxidative stress, mitochondrial dysfunction and inflammation, and these beneficial effects were attributed to the activation of AMPK signaling pathway and suppression of NF-κB signaling pathway. CONCLUSION POF could be exploited as a promising phytotherapy in the treatment of NAFLD.
Collapse
Affiliation(s)
- Kai Chen
- Department of Pharmacy, The Hospital Affiliated to Medical School of Yangzhou University (Taizhou People's Hospital), Taizhou, China
| | - Jianjiang Qu
- Department of Pharmacy, The Hospital Affiliated to Medical School of Yangzhou University (Taizhou People's Hospital), Taizhou, China
| | - Hongwei Chen
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Jue Wang
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Huilian Hua
- Department of Pharmacy, The Hospital Affiliated to Medical School of Yangzhou University (Taizhou People's Hospital), Taizhou, China
| | - Jindong Li
- Department of Pharmacy, The Hospital Affiliated to Medical School of Yangzhou University (Taizhou People's Hospital), Taizhou, China
| | - Lei Zhou
- Jiangsu Engineering Research Center of Cardiovascular Drugs Targeting Endothelial Cells, College of Health Sciences, School of Life Science, Jiangsu Normal University, Xuzhou, China
| | - Wei Zhang
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China.
| | - Zheng Li
- Jiangsu Engineering Research Center of Cardiovascular Drugs Targeting Endothelial Cells, College of Health Sciences, School of Life Science, Jiangsu Normal University, Xuzhou, China; State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China.
| |
Collapse
|
233
|
Di T, Zhai C, Zhao J, Wang Y, Chen Z, Li P. Taxifolin inhibits keratinocyte proliferation and ameliorates imiquimod-induced psoriasis-like mouse model via regulating cytoplasmic phospholipase A2 and PPAR-γ pathway. Int Immunopharmacol 2021; 99:107900. [PMID: 34233233 DOI: 10.1016/j.intimp.2021.107900] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/01/2021] [Accepted: 06/15/2021] [Indexed: 12/29/2022]
Abstract
Psoriasis is a skin disease with autoimmune tendency, and taxifolin is an effective flavonoid with anti-inflammatory activity. It has been reported that taxifolin alleviates psoriatic dermatitis, but the detailed regulatory mechanism of keratinocyte proliferation is unclear. In this study, we revealed the mechanism of taxifolin on imiquimod-induced inflammatory infiltration and keratinocyte over-proliferation. Our results show that taxifolin prevented proliferation cycle of keratinocyte in a concentration-dependent manner. Over-proliferation and abnormal apoptosis of epidermal cells were obvious in the mouse model of psoriasis induced by imiquimod. Taxifolin treatment improved erythema and scales of psoriatic lesions in mice, and reduced the proportion of CD3 + cells, especially γδT cells, in lesions and thymus. Therefore, taxifolin decreased the expression level of IL-17A-dominated inflammatory cytokines. Proteomic analysis showed that 30 up-regulated proteins and 23 down-regulated proteins were compared with the lesions before and after the treatment with taxifolin. Among them, cytoplasmic phospholipase A2 (cPLA2), the key enzyme of the pro-inflammatory mediator, was the most significantly down-regulated protein. And enriched KEGG pathway shown that PPAR-γ pathway was most involved. Taxifolin significantly reduced p-cPLA2 and increased PPAR-γ protein level in keratinocytes and lesions induced by IL-17 and imiquimod respectively. Meanwhile, phosphorylation of ERK and P-38 were also inhibited. These results suggest that taxifolin prevented imiquimode-induced excessive immune activation and keratinocyte proliferation by decreasing p-cPLA2 and regulating the PPAR-γ pathway. Our study provides new insights into the cellular regulatory mechanisms of taxifolin in psoriasis.
Collapse
Affiliation(s)
- Tingting Di
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing, China
| | - Chunyan Zhai
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing, China
| | - Jingxia Zhao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing, China
| | - Yan Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing, China
| | - Zhaoxia Chen
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing, China
| | - Ping Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing, China.
| |
Collapse
|
234
|
Akbari R, Behdarvand T, Afarin R, Yaghooti H, Jalali MT, Mohammadtaghvaei N. Saroglitazar improved hepatic steatosis and fibrosis by modulating inflammatory cytokines and adiponectin in an animal model of non-alcoholic steatohepatitis. BMC Pharmacol Toxicol 2021; 22:53. [PMID: 34593018 PMCID: PMC8485507 DOI: 10.1186/s40360-021-00524-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/20/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) have become significant global health concerns. In the present study, we aimed to investigate the effects of saroglitazar, a dual PPARα/γ agonist, fenofibrate, a PPAR-α agonist, and pioglitazone, a PPAR-γ agonist on an animal model of NASH. METHODS Male Wistar rats were fed a high-fat (HF) emulsion via gavage for 7 weeks to induce NASH. The HF-treated rats were grouped into four groups to receive saroglitazar, pioglitazone, fenofibrate, or vehicle. We measured body and liver weight, liver enzymes, serum levels of adiponectin and leptin. We also performed histopathological examinations and gene expression analysis of interleukin 6 (IL-6), tumor necrosis factor-alpha (TNF- α), transforming growth factor-beta (TGF-β), and monocyte chemoattractant protein 1 (MCP-1). RESULTS Body weight was markedly normalized by both saroglitazar and fenofibrate, while the liver index only decreased significantly with saroglitazar. Saroglitazar corrected ALT, AST, leptin, and adiponectin levels better than pioglitazone and fenofibrate. All PPAR agonists significantly attenuated the upregulation of the proinflammatory and TGF-β genes, which correlated with the improved steatosis, inflammation of liver tissue, and fibrotic lesions. CONCLUSIONS As documented by our results, the dual activation of PPARα/γ by saroglitazar could effectively improve steatosis, fibrosis, and aspects of necro-inflammation in the HF-induced NASH model more than fenofibrate and pioglitazone, and it can be more beneficial in the management of NASH.
Collapse
Affiliation(s)
- Rasoul Akbari
- Hyperlipidemia Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Laboratory Sciences, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Tahereh Behdarvand
- Hyperlipidemia Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Laboratory Sciences, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Reza Afarin
- Hyperlipidemia Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Laboratory Sciences, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hamid Yaghooti
- Hyperlipidemia Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Taha Jalali
- Hyperlipidemia Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Laboratory Sciences, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Narges Mohammadtaghvaei
- Hyperlipidemia Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Department of Laboratory Sciences, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
235
|
Boutot ME, Whitcomb BW, Abdelouahab N, Baccarelli AA, Boivin A, Caku A, Gillet V, Martinez G, Pasquier JC, Zhu J, Takser L, St-Cyr L, Suvorov A. In Utero Exposure to Persistent Organic Pollutants and Childhood Lipid Levels. Metabolites 2021; 11:657. [PMID: 34677372 PMCID: PMC8540619 DOI: 10.3390/metabo11100657] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/21/2021] [Accepted: 09/24/2021] [Indexed: 12/15/2022] Open
Abstract
Animal studies have shown that developmental exposures to polybrominated diphenyl ethers (PBDE) permanently affect blood/liver balance of lipids. No human study has evaluated associations between in utero exposures to persistent organic pollutants (POPs) and later life lipid metabolism. In this pilot, maternal plasma levels of PBDEs (BDE-47, BDE-99, BDE-100, and BDE-153) and polychlorinated biphenyls (PCB-138, PCB-153, and PCB-180) were determined at delivery in participants of GESTation and Environment (GESTE) cohort. Total cholesterol (TCh), triglycerides (TG), low- and high-density lipoproteins (LDL-C and HDL-C), total lipids (TL), and PBDEs were determined in serum of 147 children at ages 6-7. General linear regression was used to estimate the relationship between maternal POPs and child lipid levels with adjustment for potential confounders, and adjustment for childhood POPs. In utero BDE-99 was associated with lower childhood levels of TG (p = 0.003), and non-significantly with HDL-C (p = 0.06) and TL (p = 0.07). Maternal PCB-138 was associated with lower childhood levels of TG (p = 0.04), LDL-C (p = 0.04), and TL (p = 0.02). Our data indicate that in utero exposures to POPs may be associated with long lasting decrease in circulating lipids in children, suggesting increased lipid accumulation in the liver, a mechanism involved in NAFLD development, consistent with previously reported animal data.
Collapse
Affiliation(s)
- Maegan E. Boutot
- Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA; (M.E.B.); (B.W.W.)
| | - Brian W. Whitcomb
- Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA; (M.E.B.); (B.W.W.)
| | - Nadia Abdelouahab
- Department of Obstetrics and Gynecology, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, QC J1H 5N4, Canada; (N.A.); (J.-C.P.)
| | - Andrea A. Baccarelli
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY 10032, USA;
| | - Amélie Boivin
- Department of Pediatrics, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, QC J1H 5N4, Canada; (A.B.); (V.G.); (L.S.-C.)
| | - Artuela Caku
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, QC J1H 5N4, Canada;
| | - Virginie Gillet
- Department of Pediatrics, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, QC J1H 5N4, Canada; (A.B.); (V.G.); (L.S.-C.)
| | - Guillaume Martinez
- Department of Chemistry, Faculty of Sciences, Sherbrooke, QC J1K 2R1, Canada;
| | - Jean-Charles Pasquier
- Department of Obstetrics and Gynecology, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, QC J1H 5N4, Canada; (N.A.); (J.-C.P.)
| | - Jiping Zhu
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON K1A 0K9, Canada;
| | - Larissa Takser
- Department of Pediatrics & Department of Psychiatry, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, QC J1H 5N4, Canada;
| | - Lindsay St-Cyr
- Department of Pediatrics, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, QC J1H 5N4, Canada; (A.B.); (V.G.); (L.S.-C.)
| | - Alexander Suvorov
- Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA; (M.E.B.); (B.W.W.)
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA
| |
Collapse
|
236
|
Zhu J, Wu M, Zhou H, Cheng L, Wei X, Wang Y. Liubao brick tea activates the PI3K-Akt signaling pathway to lower blood glucose, metabolic disorders and insulin resistance via altering the intestinal flora. Food Res Int 2021; 148:110594. [PMID: 34507739 DOI: 10.1016/j.foodres.2021.110594] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/02/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022]
Abstract
Diabetes and its related metabolic disorders are worldwide public health issues. Many studies have shown that changes in the structure and composition of the intestinal flora are closely related to the host's physiological and pathological processes. In this study, we aim to explore the effect of Liubao tea (LBT) extract on hyperglycemic mice with metabolic disorders and intestinal flora dysbiosis and to further study its regulatory effect on insulin resistance and its potential regulatory mechanism. Our results show that LBT had a good hypoglycemic effect and could significantly alleviate the metabolic disorder evoked by hyperglycemia. The gut microbial sequencing showed that LBT treatment increased the diversity of intestinal flora, increased the abundance of beneficial bacteria, and reduced the abundance of harmful or conditional pathogenic bacteria, as well as significantly altered 39 of the top 50 OTUs with abundance. Besides, LBT could activate the PI3K-Akt-PPARs-GLUT2 cascade signaling pathway to improve metabolic disorders, thereby alleviating insulin resistance. These results suggest that LBT has excellent potential to become a natural functional food for the prevention of hyperglycemia and insulin resistance.
Collapse
Affiliation(s)
- Jiangxiong Zhu
- Institute of Food Engineering, College of Life Sciences, Shanghai Normal University, 100 Guilin Road, Xuhui District, Shanghai 200234, China
| | - Meirong Wu
- Institute of Food Engineering, College of Life Sciences, Shanghai Normal University, 100 Guilin Road, Xuhui District, Shanghai 200234, China
| | - Hui Zhou
- Institute of Food Engineering, College of Life Sciences, Shanghai Normal University, 100 Guilin Road, Xuhui District, Shanghai 200234, China
| | - Lizeng Cheng
- Department of Food Science & Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| | - Xinlin Wei
- Institute of Food Engineering, College of Life Sciences, Shanghai Normal University, 100 Guilin Road, Xuhui District, Shanghai 200234, China; Department of Food Science & Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China.
| | - Yuanfeng Wang
- Institute of Food Engineering, College of Life Sciences, Shanghai Normal University, 100 Guilin Road, Xuhui District, Shanghai 200234, China.
| |
Collapse
|
237
|
Ma L, Lian Y, Tang J, Chen F, Gao H, Zhou Z, Hou N, Yi W. Identification of the anti-fungal drug fenticonazole nitrate as a novel PPARγ-modulating ligand with good therapeutic index: Structure-based screening and biological validation. Pharmacol Res 2021; 173:105860. [PMID: 34461220 DOI: 10.1016/j.phrs.2021.105860] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 08/16/2021] [Accepted: 08/25/2021] [Indexed: 10/20/2022]
Abstract
In this study, SB-VHTS of the old drug library was conducted to seek for novel PPARγ ligand. In the end, an antifungal drug, FN, was identified in vitro and in vivo as a new and potent PPARγ-modulating ligand to demonstrate significantly anti-diabetic and anti-NAFLD efficacies with minimized side effects induced by PPARγ full agonists TZDs drugs. Further mechanistic investigations revealed that FN showed such desired pharmacological properties mainly through selectively activating the expressions of Adiponectin and GLUT4, effectively promoting the Akt Ser473 phosphorylation, inhibiting the expressions of proinflammatory genes including TNF-α, IL-1β and IL-6 and blocking the PPARγ Ser273 phosphorylation mediated by CDK5 without leading to adipogenesis and increasing the expressions of key adipogenic genes CD36, AP2, LPL, C/EBPα, FASN and PPARγ. Subsequently, a molecular docking study revealed an interesting binding mode between FN and PPARγ LBD including the hydrogen-bonding network among oxygen atom, sulfur atom and nitrogen atom in FN respectively with the PPARγ residues Cys285, Tyr327 and Ser342, which gave proof of concept for the above anti-diabetic action mechanism. Taken together, our findings not only suggest that FN can serve as the new, safe and highly efficacious anti-diabetic and anti-NAFLD agents for clinical use, they can also provide a molecular basis for the future development of PPARγ modulators with a high therapeutic index and the possibility to explore new uses of old drugs for immediate drug discovery.
Collapse
Affiliation(s)
- 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
| | - Yuling Lian
- 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
| | - Junyuan Tang
- 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
| | - 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
| | - Hui Gao
- 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.
| | - Ning Hou
- 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
|
238
|
Žiberna L, Jenko-Pražnikar Z, Petelin A. Serum Bilirubin Levels in Overweight and Obese Individuals: The Importance of Anti-Inflammatory and Antioxidant Responses. Antioxidants (Basel) 2021; 10:antiox10091352. [PMID: 34572984 PMCID: PMC8472302 DOI: 10.3390/antiox10091352] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 12/14/2022] Open
Abstract
Obesity is a chronic condition involving low-grade inflammation and increased oxidative stress; thus, obese and overweight people have lower values of serum bilirubin. Essentially, bilirubin is a potent endogenous antioxidant molecule with anti-inflammatory, immunomodulatory, antithrombotic, and endocrine properties. This review paper presents the interplay between obesity-related pathological processes and bilirubin, with a focus on adipose tissue and adipokines. We discuss potential strategies to mildly increase serum bilirubin levels in obese patients as an adjunctive therapeutic approach.
Collapse
Affiliation(s)
- Lovro Žiberna
- Institute of Pharmacology and Experimental Toxicology, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia;
| | | | - Ana Petelin
- Faculty of Health Sciences, University of Primorska, SI-6310 Izola, Slovenia;
- Correspondence: ; Tel.: +386-5-66-2469
| |
Collapse
|
239
|
Ji L, Song W, Fang H, Li W, Geng J, Wang Y, Guo L, Cai H, Yang T, Li H, Yang G, Li Q, Liu K, Li S, Liu Y, Shi F, Li X, Gao X, Tian H, Ji Q, Su Q, Zhou Z, Wang W, Zhou Z, Li X, Xu Y, Ning Z, Cao H, Pan D, Yao H, Lu X, Jia W. Efficacy and safety of chiglitazar, a novel peroxisome proliferator-activated receptor pan-agonist, in patients with type 2 diabetes: a randomized, double-blind, placebo-controlled, phase 3 trial (CMAP). Sci Bull (Beijing) 2021; 66:1571-1580. [PMID: 36654286 DOI: 10.1016/j.scib.2021.03.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 09/21/2020] [Accepted: 03/09/2021] [Indexed: 02/03/2023]
Abstract
Chiglitazar (Carfloglitazar) is a novel non-thiazolidinedione (TZD) structured peroxisome proliferator-activated receptor (PPAR) pan-agonist that has shown promising effects on glycemic control and lipid regulation in patients with type 2 diabetes in previous clinical studies. This randomized phase 3 trial aimed to compare the efficacy and safety of chiglitazar with placebo in patients with type 2 diabetes with insufficient glycemic control by strict diet and exercise alone. Eligible patients were randomly assigned to receive chiglitazar 32 mg (n = 167), chiglitazar 48 mg (n = 166), or placebo (n = 202) once daily. The primary endpoint was the change in glycosylated hemoglobin A1c (HbA1c) at week 24 with superiority of chiglitazar over placebo. The results showed that both chiglitazar 32 and 48 mg resulted in significant and clinically meaningful reductions in HbA1c, and placebo-adjusted estimated treatment differences at week 24 for chiglitazar 32 and 48 mg were -0.87% (95% confidential interval (CI): -1.10 to -0.65; P < 0.0001) and -1.05% (95% CI: -1.29 to -0.81; P < 0.0001), respectively. Secondary efficacy parameters including glycemic control, insulin sensitivity and triglyceride reduction were also significantly improved in the chiglitazar groups. The overall frequency of adverse events and study discontinuation attributable to adverse events were similar among the groups. Low incidences of mild edema and body weight gain were reported in the chiglitazar dose groups. The results from this phase 3 trial demonstrated that the PPAR pan-agonist chiglitazar possesses an overall good efficacy and safety profile in patients with type 2 diabetes inadequately controlled with lifestyle interventions, thereby providing adequate supporting evidence for using this PPAR pan-agonist as a treatment option for type 2 diabetes.
Collapse
Affiliation(s)
- Linong Ji
- Peking University People's Hospital, Beijing 100044, China.
| | - Weihong Song
- Chenzhou No.1 People's Hospital, Chenzhou 423000, China
| | - Hui Fang
- Tangshan Gongren Hospital, Tangshan 063000, China
| | - Wei Li
- The Affiliated Hospital of Xuzhou Medical College, Xuzhou 221006, China
| | - Jianlin Geng
- Harrison International Peace Hospital, Hengshui 053000, China
| | - Yangang Wang
- The Affiliate Hospital of Qingdao University, Qingdao 266003, China
| | - Lian Guo
- Chongqing Three Gorges Central Hospital, Chongqing 404000, China
| | - Hanqing Cai
- The Second Hospital of Jilin University, Changchun 130041, China
| | - Tao Yang
- Jiangsu Province Hospital, Nanjing 210029, China
| | - Hongmei Li
- China Meitan General Hospital, Beijing 100028, China
| | - Gangyi Yang
- The Second Affiliate Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Qifu Li
- The First Affiliate Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Kuanzhi Liu
- The Third Hospital of Hebei Medical University, Shijiazhuang 050051, China
| | - Shuying Li
- Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yanjun Liu
- The 306th Hospital of PLA, Beijing 100101, China
| | - Fuyan Shi
- Baogang Hospital of Inner Mongolia, Baotou 014010, China
| | - Xinsheng Li
- Cangzhou's Central Hospital, Cangzhou 031706, China
| | - Xin Gao
- Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Haoming Tian
- Huaxi Hopsital of Sichuan University, Chengdu 610041, China
| | - Qiuhe Ji
- The First Affiliated Hospital of The 4th Military Medical University, Xi'an 710000, China
| | - Qing Su
- Xin Hua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Zhiguang Zhou
- The Second Xiangya Hospital of Central South University, Changsha 410008, China
| | - Wenbo Wang
- Peking University Shougang Hospital, Beijing 100144, China
| | - Zunhai Zhou
- The Central Hospital of Yangpu District of Shanghai, Shanghai 200090, China
| | - Xuejun Li
- The First Affiliate Hospital of Xiamen University, Xiamen 361003, China
| | - Yancheng Xu
- Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Zhiqiang Ning
- Shenzhen Chipscreen Biosciences Co., Ltd., Shenzhen 518057, China
| | - Haixiang Cao
- Shenzhen Chipscreen Biosciences Co., Ltd., Shenzhen 518057, China
| | - Desi Pan
- Shenzhen Chipscreen Biosciences Co., Ltd., Shenzhen 518057, China
| | - He Yao
- Shenzhen Chipscreen Biosciences Co., Ltd., Shenzhen 518057, China
| | - Xianping Lu
- Shenzhen Chipscreen Biosciences Co., Ltd., Shenzhen 518057, China
| | - Weiping Jia
- Shanghai 6th People's Hospital, Shanghai 200233, China.
| |
Collapse
|
240
|
Jia W, Ma J, Miao H, Wang C, Wang X, Li Q, Lu W, Yang J, Zhang L, Yang J, Wang G, Zhang X, Zhang M, Sun L, Yu X, Du J, Shi B, Xiao C, Zhu D, Liu H, Zhong L, Xu C, Xu Q, Liang G, Zhang Y, Li G, Gu M, Liu J, Yuan G, Yan Z, Yan D, Ye S, Zhang F, Ning Z, Cao H, Pan D, Yao H, Lu X, Ji L. Chiglitazar monotherapy with sitagliptin as an active comparator in patients with type 2 diabetes: a randomized, double-blind, phase 3 trial (CMAS). Sci Bull (Beijing) 2021; 66:1581-1590. [PMID: 36654287 DOI: 10.1016/j.scib.2021.02.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 09/13/2020] [Accepted: 02/05/2021] [Indexed: 02/03/2023]
Abstract
Chiglitazar (Carfloglitazar) is a novel peroxisome proliferator-activated receptor (PPAR) pan-agonist that has shown promising effects on glycemic control and lipid regulation in patients with type 2 diabetes. In this randomized phase 3 trial, we compared the efficacy and safety of chiglitazar with sitagliptin in patients with type 2 diabetes who had insufficient glycemic control despite a strict diet and exercise regimen. Eligible patients were randomized (1:1:1) to receive chiglitazar 32 mg (n = 245), chiglitazar 48 mg (n = 246), or sitagliptin 100 mg (n = 248) once daily for 24 weeks. The primary endpoint was the change in glycosylated hemoglobin A1C (HbA1c) from baseline at week 24 with the non-inferiority of chiglitazar over sitagliptin. Both chiglitazar and sitagliptin significantly reduced HbA1c at week 24 with values of -1.40%, -1.47%, and -1.39% for chiglitazar 32 mg, chiglitazar 48 mg, and sitagliptin 100 mg, respectively. Chiglitazar 32 and 48 mg were both non-inferior to sitagliptin 100 mg, with mean differences of -0.04% (95% confidential interval (CI) -0.22 to 0.15) and -0.08% (95% CI -0.27 to 0.10), respectively. Compared with sitagliptin, greater reduction in fasting and 2-h postprandial plasma glucose and fasting insulin was observed with chiglitazar. Overall adverse event rates were similar between the groups. A small increase in mild edema in the chiglitazar 48 mg group and slight weight gain in both chiglitazar groups were reported. The overall results demonstrated that chiglitazar possesses good efficacy and safety profile in patients with type 2 diabetes inadequately controlled with lifestyle interventions, thereby providing adequate supporting evidence for using this PPAR pan-agonist as a treatment option for type 2 diabetes.
Collapse
Affiliation(s)
- Weiping Jia
- Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Disease, Shanghai 200233, China.
| | - Jianhua Ma
- Nanjing First Hospital, Nanjing 210029, China
| | - Heng Miao
- The Second Hospital Affiliated to Nanjing Medical University, Nanjing 210011, China
| | - Changjiang Wang
- The First Hospital Affiliated to Anhui Medical University, Hefei 230031, China
| | - Xiaoyue Wang
- The First People's Hospital of Yueyang, Yueyang 414000, China
| | - Quanmin Li
- PLA Rocket Force Characteristic Medical Center, Beijing 100085, China
| | - Weiping Lu
- Huai'an First People's Hospital, Huai'an 223300, China
| | - Jialin Yang
- The Central Hospital of Minhang District of Shanghai, Shanghai 201100, China
| | - Lihui Zhang
- The Second Hospital of Heibei Medical University, Shijiazhuang 050000, China
| | - Jinkui Yang
- Beijing Tongren Hospital Affiliated to Capital Medical University, Beijing 100730, China
| | - Guixia Wang
- The First Hospital of Jilin University, Changchun 130021, China
| | - Xiuzhen Zhang
- Tongji Hospital of Tongji University, Shanghai 200092, China
| | - Min Zhang
- The Qingpu Branch of Zhongshan Hospital Affiliate to Fudan University, Shanghai 201700, China
| | - Li Sun
- Siping Central People's Hospital, Siping 136000, China
| | - Xuefeng Yu
- Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jianling Du
- The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Bingyin Shi
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Changqing Xiao
- The First Affiliated Hospital of Guangxi Medical University (The Western Hospital), Nanning 530021, China
| | - Dalong Zhu
- Gulou Hospital Affiliated to Nanjing Medical University, Nanjing 210008, China
| | - Hong Liu
- The First Affiliated Hospital of Guangxi Medical University (The Eastern Hospital), Nanning 530021, China
| | - Liyong Zhong
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Chun Xu
- The General Hospital of the Chinese People's Armed Police Forces, Beijing 100022, China
| | - Qi Xu
- The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | | | - Ying Zhang
- The Third Hospital Affiliated to Guangzhou Medical College, Guangzhou 510150, China
| | | | - Mingyu Gu
- Shanghai First People's Hospital, Shanghai 200080, China
| | - Jun Liu
- Shanghai 5th People's Hospital, Shanghai 200040, China
| | - Guoyue Yuan
- The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Zhaoli Yan
- The Affiliated Hospital of Inner Mongolia, Hohhot 000306, China
| | - Dewen Yan
- Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Shandong Ye
- Anhui Provincial Hospital, Hefei 518035, China
| | - Fan Zhang
- Beijing University Shenzhen Hospital, Shenzhen 518036, China
| | - Zhiqiang Ning
- Shenzhen Chipscreen Biosciences, Ltd., Shenzhen 518057, China
| | - Haixiang Cao
- Shenzhen Chipscreen Biosciences, Ltd., Shenzhen 518057, China
| | - Desi Pan
- Shenzhen Chipscreen Biosciences, Ltd., Shenzhen 518057, China
| | - He Yao
- Shenzhen Chipscreen Biosciences, Ltd., Shenzhen 518057, China
| | - Xianping Lu
- Shenzhen Chipscreen Biosciences, Ltd., Shenzhen 518057, China
| | - Linong Ji
- Peking University People's Hospital, Beijing 100044, China.
| |
Collapse
|
241
|
Zhao GN, Tian ZW, Tian T, Zhu ZP, Zhao WJ, Tian H, Cheng X, Hu FJ, Hu ML, Tian S, Ding T, Chen S, Ji YX, Zhang P, Zhang XJ, She ZG, Yuan Y, Chen W, Bai L, Li H. TMBIM1 is an inhibitor of adipogenesis and its depletion promotes adipocyte hyperplasia and improves obesity-related metabolic disease. Cell Metab 2021; 33:1640-1654.e8. [PMID: 34107313 DOI: 10.1016/j.cmet.2021.05.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 04/12/2021] [Accepted: 05/13/2021] [Indexed: 01/09/2023]
Abstract
Obesity is characterized by the excessive accumulation of the white adipose tissue (WAT), but healthy expansion of WAT via adipocyte hyperplasia can offset the negative metabolic effects of obesity. Thus, identification of novel adipogenesis regulators that promote hyperplasia may lead to effective therapies for obesity-induced metabolic disorders. Using transcriptomic approaches, we identified transmembrane BAX inhibitor motif-containing 1 (TMBIM1) as an inhibitor of adipogenesis. Gain or loss of function of TMBIM1 in preadipocytes inhibited or promoted adipogenesis, respectively. In vivo, in response to caloric excess, adipocyte precursor (AP)-specific Tmbim1 knockout (KO) mice displayed WAT hyperplasia and improved systemic metabolic health, while overexpression of Tmbim1 in transgenic mice showed the opposite effects. Moreover, mature adipocyte-specific Tmbim1 KO did not affect WAT cellularity or nutrient homeostasis. Mechanistically, TMBIM1 binds to and promotes the autoubiquitination and degradation of NEDD4, which is an E3 ligase that stabilizes PPARγ. Our data show that TMBIM1 is a potent repressor of adipogenesis and a potential therapeutic target for obesity-related metabolic disease.
Collapse
Affiliation(s)
- Guang-Nian Zhao
- Medical Science Research Center, Zhongnan Hospital, School of Basic Medical Sciences, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Zheng-Wei Tian
- Medical Science Research Center, Zhongnan Hospital, School of Basic Medical Sciences, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Tian Tian
- Institute of Model Animal, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhi-Peng Zhu
- Medical Science Research Center, Zhongnan Hospital, School of Basic Medical Sciences, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Wen-Jie Zhao
- Medical Science Research Center, Zhongnan Hospital, School of Basic Medical Sciences, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Han Tian
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Xu Cheng
- Institute of Model Animal, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Feng-Jiao Hu
- Medical Science Research Center, Zhongnan Hospital, School of Basic Medical Sciences, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Man-Li Hu
- Medical Science Research Center, Zhongnan Hospital, School of Basic Medical Sciences, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Song Tian
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Ting Ding
- Department of Endocrinology, Huanggang Central Hospital, Huanggang, China; Huanggang Institute of Translational Medicine, Huanggang, China
| | - Siping Chen
- Department of Endocrinology, Huanggang Central Hospital, Huanggang, China; Huanggang Institute of Translational Medicine, Huanggang, China
| | - Yan-Xiao Ji
- Medical Science Research Center, Zhongnan Hospital, School of Basic Medical Sciences, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Peng Zhang
- Medical Science Research Center, Zhongnan Hospital, School of Basic Medical Sciences, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Xiao-Jing Zhang
- Medical Science Research Center, Zhongnan Hospital, School of Basic Medical Sciences, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Zhi-Gang She
- Institute of Model Animal, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yufeng Yuan
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China; Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Hubei, China.
| | - Wenping Chen
- Department of Endocrinology, Huanggang Central Hospital, Huanggang, China; Huanggang Institute of Translational Medicine, Huanggang, China.
| | - Lan Bai
- Institute of Model Animal, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Hongliang Li
- Medical Science Research Center, Zhongnan Hospital, School of Basic Medical Sciences, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Huanggang Institute of Translational Medicine, Huanggang, China.
| |
Collapse
|
242
|
Ferguson D, Finck BN. Emerging therapeutic approaches for the treatment of NAFLD and type 2 diabetes mellitus. Nat Rev Endocrinol 2021; 17:484-495. [PMID: 34131333 PMCID: PMC8570106 DOI: 10.1038/s41574-021-00507-z] [Citation(s) in RCA: 246] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/29/2021] [Indexed: 12/15/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has emerged as the most prevalent liver disease in the world, yet there are still no approved pharmacological therapies to prevent or treat this condition. NAFLD encompasses a spectrum of severity, ranging from simple steatosis to non-alcoholic steatohepatitis (NASH). Although NASH is linked to an increased risk of hepatocellular carcinoma and cirrhosis and has now become the leading cause of liver failure-related transplantation, the majority of patients with NASH will ultimately die as a result of complications of type 2 diabetes mellitus (T2DM) and cardiometabolic diseases. Importantly, NAFLD is closely linked to obesity and tightly interrelated with insulin resistance and T2DM. Thus, targeting these interconnected conditions and taking a holistic attitude to the treatment of metabolic disease could prove to be a very beneficial approach. This Review will explore the latest relevant literature and discuss the ongoing therapeutic options for NAFLD focused on targeting intermediary metabolism, insulin resistance and T2DM to remedy the global health burden of these diseases.
Collapse
Affiliation(s)
- Daniel Ferguson
- Division of Geriatrics and Nutritional Sciences, Center for Human Nutrition, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Brian N Finck
- Division of Geriatrics and Nutritional Sciences, Center for Human Nutrition, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
243
|
Wang Z, Zhao J, Wang Y, Zhang T, Liu R, Chang M, Wang X. Advances in EPA-GPLs: Structural features, mechanisms of nutritional functions and sources. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.06.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
244
|
Yang Q, Zhao J, Chen D, Wang Y. E3 ubiquitin ligases: styles, structures and functions. MOLECULAR BIOMEDICINE 2021; 2:23. [PMID: 35006464 PMCID: PMC8607428 DOI: 10.1186/s43556-021-00043-2] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 04/30/2021] [Indexed: 01/10/2023] Open
Abstract
E3 ubiquitin ligases are a large family of enzymes that join in a three-enzyme ubiquitination cascade together with ubiquitin activating enzyme E1 and ubiquitin conjugating enzyme E2. E3 ubiquitin ligases play an essential role in catalyzing the ubiquitination process and transferring ubiquitin protein to attach the lysine site of targeted substrates. Importantly, ubiquitination modification is involved in almost all life activities of eukaryotes. Thus, E3 ligases might be involved in regulating various biological processes and cellular responses to stress signal associated with cancer development. Thanks to their multi-functions, E3 ligases can be a promising target of cancer therapy. A deeper understanding of the regulatory mechanisms of E3 ligases in tumorigenesis will help to find new prognostic markers and accelerate the growth of anticancer therapeutic approaches. In general, we mainly introduce the classifications of E3 ligases and their important roles in cancer progression and therapeutic functions.
Collapse
Affiliation(s)
- Quan Yang
- Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China
| | - Jinyao Zhao
- Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China
| | - Dan Chen
- Department of Pathology, First Affiliated Hospital, Dalian Medical University, Dalian, 116044, China.
| | - Yang Wang
- Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
245
|
Proteomics and metabonomics analyses of Covid-19 complications in patients with pulmonary fibrosis. Sci Rep 2021; 11:14601. [PMID: 34272434 PMCID: PMC8285535 DOI: 10.1038/s41598-021-94256-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 06/29/2021] [Indexed: 12/23/2022] Open
Abstract
Pulmonary fibrosis is a devastating disease, and the pathogenesis of this disease is not completely clear. Here, the medical records of 85 Covid-19 cases were collected, among which fibrosis and progression of fibrosis were analyzed in detail. Next, data independent acquisition (DIA) quantification proteomics and untargeted metabolomics were used to screen disease-related signaling pathways through clustering and enrichment analysis of the differential expression of proteins and metabolites. The main imaging features were lesions located in the bilateral lower lobes and involvement in five lobes. The closed association pathways were FcγR-mediated phagocytosis, PPAR signaling, TRP-inflammatory pathways, and the urea cycle. Our results provide evidence for the detection of serum biomarkers and targeted therapy in patients with Covid-19.
Collapse
|
246
|
Current and Emerging Approaches for Hepatic Fibrosis Treatment. Gastroenterol Res Pract 2021; 2021:6612892. [PMID: 34326871 PMCID: PMC8310447 DOI: 10.1155/2021/6612892] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 07/10/2021] [Indexed: 02/07/2023] Open
Abstract
Liver fibrosis resulting from chronic liver injury is a key factor to develop liver cirrhosis and risk of hepatocellular carcinoma (HCC) which are major health burden worldwide. Therefore, it is necessary for antifibrotic therapies to prevent chronic liver disease progression and HCC development. There has been tremendous progress in understanding the mechanisms of liver fibrosis in the last decade, which has created new opportunities for the treatment of this condition. In this review, we aim to make an overview on information of different potential therapies (drug treatment, cell therapy, and liver transplantation) for the liver fibrosis and hope to provide the therapeutic options available for the treatment of liver fibrosis and discuss novel approaches.
Collapse
|
247
|
Tanigawa K, Luo Y, Kawashima A, Kiriya M, Nakamura Y, Karasawa K, Suzuki K. Essential Roles of PPARs in Lipid Metabolism during Mycobacterial Infection. Int J Mol Sci 2021; 22:ijms22147597. [PMID: 34299217 PMCID: PMC8304230 DOI: 10.3390/ijms22147597] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/13/2021] [Accepted: 07/13/2021] [Indexed: 12/25/2022] Open
Abstract
The mycobacterial cell wall is composed of large amounts of lipids with varying moieties. Some mycobacteria species hijack host cells and promote lipid droplet accumulation to build the cellular environment essential for their intracellular survival. Thus, lipids are thought to be important for mycobacteria survival as well as for the invasion, parasitization, and proliferation within host cells. However, their physiological roles have not been fully elucidated. Recent studies have revealed that mycobacteria modulate the peroxisome proliferator-activated receptor (PPAR) signaling and utilize host-derived triacylglycerol (TAG) and cholesterol as both nutrient sources and evasion from the host immune system. In this review, we discuss recent findings that describe the activation of PPARs by mycobacterial infections and their role in determining the fate of bacilli by inducing lipid metabolism, anti-inflammatory function, and autophagy.
Collapse
Affiliation(s)
- Kazunari Tanigawa
- Department of Molecular Pharmaceutics, Faculty of Pharma-Science, Teikyo University, Itabashi-ku, Tokyo 173-8605, Japan; (K.T.); (Y.N.); (K.K.)
| | - Yuqian Luo
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Itabashi-ku, Tokyo 173-8605, Japan; (Y.L.); (A.K.); (M.K.)
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing 210008, China
| | - Akira Kawashima
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Itabashi-ku, Tokyo 173-8605, Japan; (Y.L.); (A.K.); (M.K.)
| | - Mitsuo Kiriya
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Itabashi-ku, Tokyo 173-8605, Japan; (Y.L.); (A.K.); (M.K.)
| | - Yasuhiro Nakamura
- Department of Molecular Pharmaceutics, Faculty of Pharma-Science, Teikyo University, Itabashi-ku, Tokyo 173-8605, Japan; (K.T.); (Y.N.); (K.K.)
| | - Ken Karasawa
- Department of Molecular Pharmaceutics, Faculty of Pharma-Science, Teikyo University, Itabashi-ku, Tokyo 173-8605, Japan; (K.T.); (Y.N.); (K.K.)
| | - Koichi Suzuki
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Itabashi-ku, Tokyo 173-8605, Japan; (Y.L.); (A.K.); (M.K.)
- Correspondence: ; Tel.: +81-3-3964-1211
| |
Collapse
|
248
|
Smati S, Canivet CM, Boursier J, Cariou B. Anti-diabetic drugs and NASH: from current options to promising perspectives. Expert Opin Investig Drugs 2021; 30:813-825. [PMID: 34214406 DOI: 10.1080/13543784.2021.1951701] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Accumulating evidence supports a bidirectional association between nonalcoholic steatohepatitis (NASH) and type 2 diabetes (T2D). There is a clinical challenge to consider pharmaceutical strategies targeting the metabolic dysfunction common to NASH and T2D pathogenesis.Areas covered: By using PubMed, we performed a literature search to review the potential beneficial effect of anti-diabetic and metabolic investigational drugs on NASH.Expert opinion: Since insulin resistance is central in the pathophysiology of both T2D and NASH, there is an urgent need for new insulin sensitizers. Peroxisome proliferator-activated receptor (PPAR) agonists, especially PPARγ and pan-PPARs agonists, have shown some beneficial effects on both NASH and liver fibrosis, but their routine use should be limited by their safety profile. Incretin-based therapies, including glucagon-like peptide 1 receptor agonists (GLP-1 RAs) and the polyagonists (GLP-1, GIP, glucagon) under development are the most promising anti-diabetic drugs for NASH treatment, mainly due to their action on body weight loss. Preliminary, preclinical and early phase studies suggest that SGLT2 inhibitors and fibroblast growth factor (FGF)19 and FGF21-based therapies are promising targets for NASH and T2D treatment. The common weakness for all of these drugs is their limited effect on liver fibrosis, potentially due to short-term trial design.
Collapse
Affiliation(s)
- Sarra Smati
- Department of Endocrinology, Université De Nantes, CHU Nantes, CNRS, INSERM, L'institut Du Thorax, Nantes, France
| | - Clémence M Canivet
- Hepato-Gastroenterology department, University Hospital, Angers, France.,HIFIH Laboratory, EA 3859, University of Angers, Angers, France
| | - Jérôme Boursier
- Hepato-Gastroenterology department, University Hospital, Angers, France.,HIFIH Laboratory, EA 3859, University of Angers, Angers, France
| | - Bertrand Cariou
- Department of Endocrinology, Université De Nantes, CHU Nantes, CNRS, INSERM, L'institut Du Thorax, Nantes, France
| |
Collapse
|
249
|
Naowaboot J, Nanna U, Chularojmontri L, Songtavisin T, Tingpej P, Sattaponpan C, Jansom C, Wattanapitayakul S. Mentha cordifolia Leaf Extract Improves Hepatic Glucose and Lipid Metabolism in Obese Mice Fed with High-Fat Diet. Prev Nutr Food Sci 2021; 26:157-165. [PMID: 34316480 PMCID: PMC8276705 DOI: 10.3746/pnf.2021.26.2.157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/18/2021] [Accepted: 03/22/2021] [Indexed: 11/21/2022] Open
Abstract
Mentha cordifolia (MC) is a popular herb used to flavor food in Thailand that exhibits several biological effects. The present study aimed to determine the role of MC in regulating glucose and lipid metabolism in mice fed a high-fat diet (HFD). ICR obese mice were fed an HFD (45 kcal% lard fat) for 12 weeks, with MC (100 and 200 mg/kg/d) treatment from Week 7. After treatment with MC for 6 weeks, mice showed significantly lower rates of hyperglycemia, hyperinsulinemia, hyperleptinemia, and hyperlipidemia, and increased amounts of serum adiponectin. Furthermore, in mice treated with MC, serum interleukin-6 and tumor necrosis factor alpha were significantly inhibited and liver histology results showed decreased lipid accumulation and liver triglyceride content vs. untreated mice. In addition, MC treatment was associated with smaller fat cells and lower gene expression of liver sterol regulatory element binding protein 1c, acetyl-CoA carboxylase, and fatty acid synthase. However, MC treatment was associated with higher carnitine palmitoyltransferase 1a gene expression and significantly higher rates of adenosine monophosphate-activated protein kinase (AMPK) phosphorylation in liver, but lower levels of phosphoenolpyruvate carboxykinase and glucose-6-phosphatase. These results indicate MC regulates glucose and lipid metabolism in a HFD-induced obese mouse model, possibly via activation of AMPK signaling pathway.
Collapse
Affiliation(s)
- Jarinyaporn Naowaboot
- Division of Pharmacology, Faculty of Medicine, Thammasat University, Pathum Thani 12120, Thailand
| | - Urarat Nanna
- Division of Pharmacology, Faculty of Medicine, Thammasat University, Pathum Thani 12120, Thailand
| | - Linda Chularojmontri
- Division of Pharmacology, Faculty of Medicine, Thammasat University, Pathum Thani 12120, Thailand
| | - Thanitsara Songtavisin
- Division of Anatomy, Faculty of Medicine, Thammasat University, Pathum Thani 12120, Thailand
| | - Pholawat Tingpej
- Division of Microbiology and Immunology, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathum Thani 12120, Thailand
| | - Chisanucha Sattaponpan
- Research Administrative Office, Faculty of Medicine, Thammasat University, Pathum Thani 12120, Thailand
| | - Chalerm Jansom
- Research Administrative Office, Faculty of Medicine, Thammasat University, Pathum Thani 12120, Thailand
| | - Suvara Wattanapitayakul
- Department of Pharmacology, Faculty of Medicine, Srinakharinwirot University, Bangkok 10110, Thailand
| |
Collapse
|
250
|
Hernández-Aguilar AI, Luciano-Villa CA, Tello-Flores VA, Beltrán-Anaya FO, Zubillaga-Guerrero MI, Flores-Alfaro E. Dysregulation of lncRNA-H19 in cardiometabolic diseases and the molecular mechanism involved : a systematic review. Expert Rev Mol Diagn 2021; 21:809-821. [PMID: 34133256 DOI: 10.1080/14737159.2021.1944808] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Cardiometabolic diseases are a global public health problem, with significant increases in their prevalence. Different epigenetic factors involved in the progression of metabolic alterations have been described, such as long non-coding RNAs (lncRNAs). H19 is a multifunctional lncRNA expressed from the maternal allele, with low expression after birth, except in the skeletal muscle and heart. Recent studies have linked its dysregulation to alterations in cell metabolism.Areas covered: H19 plays a role in the pathogenesis of coronary artery disease, nonalcoholic fatty liver disease, hepatic and renal fibrosis, insulin resistance, type 2 diabetes, and inflammation. H19 acts mainly as a competitive endogenous RNA of molecules involved in pathways that regulate cell metabolism. In this review, we analyzed the dysregulation of H19 in cardiometabolic diseases and its relationship with molecular alterations in different signaling pathways.Expert opinion: The association of H19 with the development of cardiometabolic diseases, indicates that H19 could be a therapeutic target and prognostic biomarker for these diseases. Controversies have been reported regarding the expression of H19 in some metabolic diseases, therefore, it is necessary to continue research to clarify its pathogenic effect in different organs.
Collapse
Affiliation(s)
- Ana Iris Hernández-Aguilar
- Faculty of Chemical‑Biological Sciences, Autonomous University of Guerrero, Chilpancingo, Guerrero, Mexico
| | | | | | - Fredy Omar Beltrán-Anaya
- Faculty of Chemical‑Biological Sciences, Autonomous University of Guerrero, Chilpancingo, Guerrero, Mexico
| | | | - Eugenia Flores-Alfaro
- Faculty of Chemical‑Biological Sciences, Autonomous University of Guerrero, Chilpancingo, Guerrero, Mexico
| |
Collapse
|