1
|
Yang X, Li L, Yan Y, Hu X, Li Q, Li L, Wang Y, Tao X, Yang L, Peng M, Yang J, Yang X, Gao M. Investigation of the Pharmacodynamic Components of Gastrodia elata Blume for Treatment of Type 2 Diabetes Mellitus through HPLC, Bioactivity, Network Pharmacology and Molecular Docking. Int J Mol Sci 2024; 25:10498. [PMID: 39408825 PMCID: PMC11476761 DOI: 10.3390/ijms251910498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
The occurrence of type 2 diabetes mellitus (T2DM), a worldwide chronic disease, is mainly caused by insufficient insulin production and places a huge burden on the health system. Gastrodia elata Blume (GE), a food of medicine-food homology, has been reported to have the ability to inhibit glycosidase activity, indicating its potential in the treatment of diabetes. However, the main pharmacological components of GE for the treatment of T2DM have not been fully clarified. Therefore, this study aims to clarify the pharmacological components changes of GE with different drying methods and the treatment of T2DM using HPLC, network pharmacology, molecular docking and experimental evaluations. The results showed that the GE samples processed by the steam-lyophilized method possessed the highest total content of the six marker components and the strongest antioxidant and α-glucosidase inhibitory abilities. Meanwhile, the six marker compounds had a total of 238 T2DM-related gene targets. Notably, these active compounds have good affinity for key gene targets associated with T2DM signaling pathways. In conclusion, this study revealed that different drying methods of GE affect the content of its major active compounds, antioxidant capacity, α-glucosidase inhibitory capacity and potential pharmacological effects on T2DM, indicating that it is a potential treatment of T2DM.
Collapse
Affiliation(s)
- Xiu Yang
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (X.Y.); (L.L.); (Y.Y.); (X.H.); (Q.L.); (L.L.); (Y.W.); (X.T.); (L.Y.); (M.P.); (J.Y.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550014, China
| | - Lilang Li
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (X.Y.); (L.L.); (Y.Y.); (X.H.); (Q.L.); (L.L.); (Y.W.); (X.T.); (L.Y.); (M.P.); (J.Y.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Yanfang Yan
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (X.Y.); (L.L.); (Y.Y.); (X.H.); (Q.L.); (L.L.); (Y.W.); (X.T.); (L.Y.); (M.P.); (J.Y.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Xuehao Hu
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (X.Y.); (L.L.); (Y.Y.); (X.H.); (Q.L.); (L.L.); (Y.W.); (X.T.); (L.Y.); (M.P.); (J.Y.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550014, China
| | - Qiji Li
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (X.Y.); (L.L.); (Y.Y.); (X.H.); (Q.L.); (L.L.); (Y.W.); (X.T.); (L.Y.); (M.P.); (J.Y.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Liangqun Li
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (X.Y.); (L.L.); (Y.Y.); (X.H.); (Q.L.); (L.L.); (Y.W.); (X.T.); (L.Y.); (M.P.); (J.Y.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Yu Wang
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (X.Y.); (L.L.); (Y.Y.); (X.H.); (Q.L.); (L.L.); (Y.W.); (X.T.); (L.Y.); (M.P.); (J.Y.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Xian Tao
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (X.Y.); (L.L.); (Y.Y.); (X.H.); (Q.L.); (L.L.); (Y.W.); (X.T.); (L.Y.); (M.P.); (J.Y.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Lishou Yang
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (X.Y.); (L.L.); (Y.Y.); (X.H.); (Q.L.); (L.L.); (Y.W.); (X.T.); (L.Y.); (M.P.); (J.Y.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Mei Peng
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (X.Y.); (L.L.); (Y.Y.); (X.H.); (Q.L.); (L.L.); (Y.W.); (X.T.); (L.Y.); (M.P.); (J.Y.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Juan Yang
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (X.Y.); (L.L.); (Y.Y.); (X.H.); (Q.L.); (L.L.); (Y.W.); (X.T.); (L.Y.); (M.P.); (J.Y.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Xiaosheng Yang
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (X.Y.); (L.L.); (Y.Y.); (X.H.); (Q.L.); (L.L.); (Y.W.); (X.T.); (L.Y.); (M.P.); (J.Y.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Ming Gao
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (X.Y.); (L.L.); (Y.Y.); (X.H.); (Q.L.); (L.L.); (Y.W.); (X.T.); (L.Y.); (M.P.); (J.Y.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| |
Collapse
|
2
|
Cobo-Vuilleumier N, Lorenzo PI, Martin Vazquez E, López Noriega L, Nano R, Piemonti L, Martín F, Gauthier BR. Enhancing human islet xenotransplant survival and function in diabetic immunocompetent mice through LRH-1/NR5A2 pharmacological activation. Front Immunol 2024; 15:1470881. [PMID: 39399499 PMCID: PMC11466778 DOI: 10.3389/fimmu.2024.1470881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 09/12/2024] [Indexed: 10/15/2024] Open
Abstract
The intricate etiology of type 1 diabetes mellitus (T1D), characterized by harmful interactions between the immune system and insulin-producing beta cells, has hindered the development of effective therapies including human islet transplantation, which requires strong immunosuppressants that impair beta cell survival and function. As such alternative immunomodulating therapies are required for successful transplantation. The discovery that pharmacological activation of the nuclear receptor LRH-1/NR5A2 can reverse hyperglycemia in mouse models of T1D by altering, and not suppressing the autoimmune attack, prompted us to investigate whether LRH-1/NR5A2 activation could improve human islet function/survival after xenotransplantation in immunocompetent mice. Human islets were transplanted under the kidney capsule of streptozotocin (STZ)-induced diabetic mice, and treatment with BL001 (LRH-1/NR5A2 agonist) or vehicle was administered one week post-transplant. Our study, encompassing 3 independent experiments with 3 different islet donors, revealed that mice treated for 8 weeks with BL001 exhibited lower blood glucose levels correlating with improved mouse survival rates as compared to vehicle-treated controls. Human C-peptide was detectable in BL001-treated mice at both 4 and 8 weeks indicating functional islet beta cells. Accordingly, in mice treated with BL001 for 8 weeks, the beta cell mass was preserved, while a significant decrease in alpha cells was observed compared to mice treated with BL001 for only 4 weeks. In contrast, vehicle-treated mice exhibited a reduction in insulin-expressing cells at 8 weeks compared to those at 4 weeks. These results suggest that BL001 significantly enhances the survival, engraftment, and functionality of human islets in a STZ-induced diabetic mouse model.
Collapse
Affiliation(s)
- N. Cobo-Vuilleumier
- Andalusian Center of Molecular Biology and Regenerative Medicine (CABIMER), Junta de Andalucía-University of Pablo de Olavide-University of Seville-Consejo Superior de Investigaciones Científicas (CSIC), Seville, Spain
| | - P. I. Lorenzo
- Andalusian Center of Molecular Biology and Regenerative Medicine (CABIMER), Junta de Andalucía-University of Pablo de Olavide-University of Seville-Consejo Superior de Investigaciones Científicas (CSIC), Seville, Spain
| | - E. Martin Vazquez
- Andalusian Center of Molecular Biology and Regenerative Medicine (CABIMER), Junta de Andalucía-University of Pablo de Olavide-University of Seville-Consejo Superior de Investigaciones Científicas (CSIC), Seville, Spain
| | - L. López Noriega
- Andalusian Center of Molecular Biology and Regenerative Medicine (CABIMER), Junta de Andalucía-University of Pablo de Olavide-University of Seville-Consejo Superior de Investigaciones Científicas (CSIC), Seville, Spain
| | - R. Nano
- Diabetes Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - L. Piemonti
- Diabetes Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - F. Martín
- Andalusian Center of Molecular Biology and Regenerative Medicine (CABIMER), Junta de Andalucía-University of Pablo de Olavide-University of Seville-Consejo Superior de Investigaciones Científicas (CSIC), Seville, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - B. R. Gauthier
- Andalusian Center of Molecular Biology and Regenerative Medicine (CABIMER), Junta de Andalucía-University of Pablo de Olavide-University of Seville-Consejo Superior de Investigaciones Científicas (CSIC), Seville, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| |
Collapse
|
3
|
Huang X, Wei L, Li M, Zhang Y, Kuang S, Shen Z, Liu H, Lin Z. Diabetic Macrophage Exosomal miR-381-3p Inhibits Epithelial Cell Autophagy Via NR5A2. Int Dent J 2024; 74:823-835. [PMID: 38685137 PMCID: PMC11287178 DOI: 10.1016/j.identj.2024.02.001] [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: 11/05/2023] [Revised: 01/12/2024] [Accepted: 02/02/2024] [Indexed: 05/02/2024] Open
Abstract
PURPOSE To explore the mechanism underlying autophagy disruption in gingival epithelial cells (GECs) in diabetic individuals. METHODS AND MATERIALS Bone marrow-derived macrophages (BMDMs) and GECs were extracted from C57/bl and db/db mice, the exosomes (Exo) were isolated from BMDMs. qRT‒PCR and Western blotting were performed to analyse gene expression. The AnimalTFDB database was used to identify relevant transcription factors, and miRNA sequencing was utilised to identify relevant miRNAs with the aid of the TargetScan/miRDB/miRWalk databases. A dual-luciferase assay was conducted to verify intermolecular targeting relationships. RESULTS Similar to BMDMs, BMDM-derived Exos disrupted autophagy and exerted proinflammatory effects in GEC cocultures, and ATG7 may play a vital role. AnimalTFDB database analysis and dual-luciferase assays indicated that NR5A2 is the most relevant transcription factor that regulates Atg7 expression. SiRNA-NR5A2 transfection blocked autophagy in GECs and exacerbated inflammation, whereas NR5A2 upregulation restored ATG7 expression and ameliorated ExoDM-mediated inflammation. MiRNA sequencing, with TargetScan/miRDB/miRWalk analyses and dual-luciferase assays, confirmed that miR-381-3p is the most relevant miRNA that targets NR5A2. MiR-381-3p mimic transfection blocked autophagy in GECs and exacerbated inflammation, while miR-381-3p inhibitor transfection restored ATG7 expression and attenuated ExoDM-mediated inflammation. CONCLUSION BMDM-derived Exos, which carry miR-381-3p, inhibit NR5A2 and disrupt autophagy in GECs, increasing periodontal inflammation in diabetes.
Collapse
Affiliation(s)
- Xin Huang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Linhesheng Wei
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Mengdi Li
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Yong Zhang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shuhong Kuang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zongshan Shen
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hui Liu
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhengmei Lin
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
4
|
Wu T, Lu ZF, Yu HN, Wu XS, Liu Y, Xu Y. Liver receptor homolog-1: structures, related diseases, and drug discovery. Acta Pharmacol Sin 2024; 45:1571-1581. [PMID: 38632319 PMCID: PMC11272790 DOI: 10.1038/s41401-024-01276-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 03/24/2024] [Indexed: 04/19/2024] Open
Abstract
Liver receptor homolog-1 (LRH-1), a member of the nuclear receptor superfamily, is a ligand-regulated transcription factor that plays crucial roles in metabolism, development, and immunity. Despite being classified as an 'orphan' receptor due to the ongoing debate surrounding its endogenous ligands, recent researches have demonstrated that LRH-1 can be modulated by various synthetic ligands. This highlights the potential of LRH-1 as an attractive drug target for the treatment of inflammation, metabolic disorders, and cancer. In this review, we provide an overview of the structural basis, functional activities, associated diseases, and advancements in therapeutic ligand research targeting LRH-1.
Collapse
Affiliation(s)
- Tong Wu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
- State Key Laboratory of Respiratory Disease, China-New Zealand Joint Laboratory of Biomedicine and Health, Guangdong Provincial Key Laboratory of Biocomputing, Center for Chemical Biology and Drug Discovery, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou, China
| | - Zhi-Fang Lu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
- State Key Laboratory of Respiratory Disease, China-New Zealand Joint Laboratory of Biomedicine and Health, Guangdong Provincial Key Laboratory of Biocomputing, Center for Chemical Biology and Drug Discovery, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou, China
| | - Hao-Nan Yu
- State Key Laboratory of Respiratory Disease, China-New Zealand Joint Laboratory of Biomedicine and Health, Guangdong Provincial Key Laboratory of Biocomputing, Center for Chemical Biology and Drug Discovery, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou, China
| | - Xi-Shan Wu
- State Key Laboratory of Respiratory Disease, China-New Zealand Joint Laboratory of Biomedicine and Health, Guangdong Provincial Key Laboratory of Biocomputing, Center for Chemical Biology and Drug Discovery, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou, China
| | - Yang Liu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Yong Xu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
- State Key Laboratory of Respiratory Disease, China-New Zealand Joint Laboratory of Biomedicine and Health, Guangdong Provincial Key Laboratory of Biocomputing, Center for Chemical Biology and Drug Discovery, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
5
|
Ma W, Long J, Dong L, Zhang J, Wang A, Zhang Y, Yan D. Uncovering the key pharmacodynamic material basis and possible molecular mechanism of Xiaoke formulation improve insulin resistant through a comprehensive investigation. JOURNAL OF ETHNOPHARMACOLOGY 2024; 323:117752. [PMID: 38216099 DOI: 10.1016/j.jep.2024.117752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/25/2023] [Accepted: 01/09/2024] [Indexed: 01/14/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xiaoke formulation (XKF) has been utilized in clinical practice for decades in China as a treatment option for mild to moderate type 2 diabetes. However, there is still a need for systematic research to uncover the key pharmacodynamic material basis and mechanism of XKF. AIM OF THE STUDY Aim of to investigate the distribution and metabolism of XKF in normal and insulin resistant (IR) mice were different, and elucidate its key pharmacodynamic material basis and mechanism of action. MATERIALS AND METHODS Ultra performance liquid chromatography/time of flight mass spectrometry technology was employed to investigate the differences in XKF absorption, distribution, and metabolism between normal and IR mice across blood, liver, feces, and urine samples. Further, network pharmacology was used to predict target proteins and their associated signaling pathways. Then, molecular docking was utilized to validate the activity of key pharmacodynamic components and targets. Finally, IR HepG2 cells were used to detect the glucose consumption under the action of key pharmacodynamic material basis. In addition, the expression of phosphatidylinositol 3-kinase (PI3K), protein kinase B (AKT) and phospho-protein kinase B (p-AKT) was determined using western blotting. RESULTS The study demonstrates significant distinctions in plasma and liver number and abundance of alkaloids, organic acids, flavonoids, iridoids and saponins between normal and IR mice when XKF was administered. Further analysis has shown that the representative components of XKF, including berberine, chlorogenic acid, calycosin, swertiamarin and astragaloside IV have significantly different metabolic pathways in plasma and liver. Prototypes and metabolites of these components were rarely detected in the urine and feces of mice. According to the network pharmacological analysis, these differential components are predicted to improve IR by targeting key factors such as SRC, JUN, HRAS, NOS3, FGF2, etc. Additionally, the signaling pathways involved in this process include PI3K-AKT pathway, GnRH signaling pathway, and T cell receptor signaling pathway. In addition, in vitro experiments indicate that berberine and its metabolites (berberine and demethyleneberine), chlorogenic acid and its metabolites (3-O-ferulic quinic acid and 5-O-ferulic quinic acid), calycosin and swertiamarin could improve IR in IR-HepG2 cells by elevating the expression of PI3K and AKT, leading to an increase in glucose consumption. CONCLUSION The key pharmacodynamic material basis of XKF, such as berberine and its metabolites (berberrubine and demethyleneberberine), chlorogenic acid and its metabolites (3-O-feruloylquinic acid and 5-O-feruloylquinic acid), calycosin and swertiamarin influence the glucose metabolism disorder of IR-HepG2 cells by regulating the PI3K/AKT signalling pathway, leading to an improvement in IR.
Collapse
Affiliation(s)
- Wenjuan Ma
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Benxi, Liaoning, 110016, China
| | - Jianglan Long
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Linjie Dong
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Benxi, Liaoning, 110016, China
| | - Jian Zhang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Aiting Wang
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Yu Zhang
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
| | - Dan Yan
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
| |
Collapse
|
6
|
Lambrecht R, Delgado ME, Gloe V, Schuetz K, Plazzo AP, Franke B, San Phan T, Fleming J, Mayans O, Brunner T. Liver receptor homolog-1 (NR5A2) orchestrates hepatic inflammation and TNF-induced cell death. Cell Rep 2023; 42:113513. [PMID: 38039134 DOI: 10.1016/j.celrep.2023.113513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 10/09/2023] [Accepted: 11/14/2023] [Indexed: 12/03/2023] Open
Abstract
The nuclear receptor liver receptor homolog-1 (LRH-1) has been shown to promote apoptosis resistance in various tissues and disease contexts; however, its role in liver cell death remains unexplored. Hepatocyte-specific deletion of LRH-1 causes mild steatosis and inflammation but unexpectedly shields female mice from tumor necrosis factor (TNF)-induced hepatocyte apoptosis and associated hepatitis. LRH-1-deficient hepatocytes show markedly attenuated estrogen receptor alpha and elevated nuclear factor κB (NF-κB) activity, while LRH-1 overexpression inhibits NF-κB activity. This inhibition relies on direct physical interaction of LRH-1's ligand-binding domain and the Rel homology domain of NF-κB subunit RelA. Mechanistically, increased transcription of anti-apoptotic NF-κB target genes and the proteasomal degradation of pro-apoptotic BCL-2 interacting mediator of cell death prevent mitochondrial apoptosis and ultimately protect mice from TNF-induced liver damage. Collectively, our study emphasizes LRH-1 as a critical, sex-dependent regulator of cell death and inflammation in the healthy and diseased liver.
Collapse
Affiliation(s)
- Rebekka Lambrecht
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78464 Konstanz, Germany
| | - M Eugenia Delgado
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78464 Konstanz, Germany
| | - Vincent Gloe
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78464 Konstanz, Germany
| | - Karina Schuetz
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78464 Konstanz, Germany
| | - Anna Pia Plazzo
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78464 Konstanz, Germany
| | - Barbara Franke
- Biophysics and Structural Biology, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78464 Konstanz, Germany
| | - Truong San Phan
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78464 Konstanz, Germany
| | - Jennifer Fleming
- Biophysics and Structural Biology, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78464 Konstanz, Germany
| | - Olga Mayans
- Biophysics and Structural Biology, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78464 Konstanz, Germany
| | - Thomas Brunner
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78464 Konstanz, Germany.
| |
Collapse
|
7
|
Hu J, Zhang Z, Hu H, Yang K, Zhu Z, Yang Q, Liang W. LRH-1 activation alleviates diabetes-induced podocyte injury by promoting GLS2-mediated glutaminolysis. Cell Prolif 2023; 56:e13479. [PMID: 37057309 PMCID: PMC10623971 DOI: 10.1111/cpr.13479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/25/2023] [Accepted: 03/31/2023] [Indexed: 04/15/2023] Open
Abstract
Alteration of metabolic phenotype in podocytes directly contributes to the development of albuminuria and renal injury in conditions of diabetic kidney disease (DKD). This study aimed to identify and evaluate liver receptor homologue-1 (LRH-1) as a possible therapeutic target that alleviates glutamine (Gln) metabolism disorders and mitigates podocyte injury in DKD. Metabolomic and transcriptomic analyses were performed to characterize amino acid metabolism changes in the glomeruli of diabetic mice. Next, Western blotting, immunohistochemistry assays, and immunofluorescence staining were used to detect the expression of different genes in vitro and in vivo. Furthermore, Gln and glutamate (Glu) content as well as ATP generation were examined. A decrease in LRH-1 and glutaminase 2 (GLS2) expression was detected in diabetic podocytes. Conversely, the administration of LRH-1 agonist (DLPC) upregulated the expression of GLS2 and promoted glutaminolysis, with an improvement in mitochondrial dysfunction and less apoptosis in podocytes compared to those in vehicle-treated db/db mice. Our study indicates the essential role of LRH-1 in governing the Gln metabolism of podocytes, targeting LRH-1 could restore podocytes from diabetes-induced disturbed glutaminolysis in mitochondria.
Collapse
Affiliation(s)
- Jijia Hu
- Division of NephrologyRenmin Hospital of Wuhan UniversityWuhanHubeiChina
- Nephrology and Urology Research Institute of Wuhan UniversityWuhanHubeiChina
| | - Zongwei Zhang
- Division of NephrologyRenmin Hospital of Wuhan UniversityWuhanHubeiChina
- Nephrology and Urology Research Institute of Wuhan UniversityWuhanHubeiChina
| | - Hongtu Hu
- Division of NephrologyRenmin Hospital of Wuhan UniversityWuhanHubeiChina
| | - Keju Yang
- Division of NephrologyRenmin Hospital of Wuhan UniversityWuhanHubeiChina
- The First College of Clinical Medical Science, China Three Gorges UniversityYichangHubeiChina
| | - Zijing Zhu
- Division of NephrologyRenmin Hospital of Wuhan UniversityWuhanHubeiChina
| | - Qian Yang
- Division of NephrologyRenmin Hospital of Wuhan UniversityWuhanHubeiChina
- Nephrology and Urology Research Institute of Wuhan UniversityWuhanHubeiChina
| | - Wei Liang
- Division of NephrologyRenmin Hospital of Wuhan UniversityWuhanHubeiChina
- Nephrology and Urology Research Institute of Wuhan UniversityWuhanHubeiChina
| |
Collapse
|
8
|
Martín-Vázquez E, Cobo-Vuilleumier N, López-Noriega L, Lorenzo PI, Gauthier BR. The PTGS2/COX2-PGE 2 signaling cascade in inflammation: Pro or anti? A case study with type 1 diabetes mellitus. Int J Biol Sci 2023; 19:4157-4165. [PMID: 37705740 PMCID: PMC10496497 DOI: 10.7150/ijbs.86492] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/24/2023] [Indexed: 09/15/2023] Open
Abstract
Prostaglandins are lipid mediators involved in physiological processes, such as constriction or dilation of blood vessels, but also pathophysiological processes, which include inflammation, pain and fever. They are produced by almost all cell types in the organism by activation of Prostaglandin endoperoxide synthases/Cyclooxygenases. The inducible Prostaglandin Endoperoxide Synthase 2/Cyclooxygenase 2 (PTGS2/COX2) plays an important role in pathologies associated with inflammatory signaling. The main product derived from PTGS2/COX2 expression and activation is Prostaglandin E2 (PGE2), which promotes a wide variety of tissue-specific effects, pending environmental inputs. One of the major sources of PGE2 are infiltrating inflammatory cells - the production of this molecule increases drastically in damaged tissues. Immune infiltration is a hallmark of type 1 diabetes mellitus, a multifactorial disease that leads to autoimmune-mediated pancreatic beta cell destruction. Controversial effects for the PTGS2/COX2-PGE2 signaling cascade in pancreatic islet cells subjected to diabetogenic conditions have been reported, allocating PGE2 as both, cause and consequence of inflammation. Herein, we review the main effects of this molecular pathway in a tissue-specific manner, with a special emphasis on beta cell mass protection/destruction and its potential role in the prevention or development of T1DM. We also discuss strategies to target this pathway for future therapies.
Collapse
Affiliation(s)
- Eugenia Martín-Vázquez
- Andalusian Center of Molecular Biology and Regenerative Medicine CABIMER, Junta de Andalucia-University of Pablo de Olavide-University of Seville-CSIC, Seville, Spain
| | - Nadia Cobo-Vuilleumier
- Andalusian Center of Molecular Biology and Regenerative Medicine CABIMER, Junta de Andalucia-University of Pablo de Olavide-University of Seville-CSIC, Seville, Spain
| | - Livia López-Noriega
- Andalusian Center of Molecular Biology and Regenerative Medicine CABIMER, Junta de Andalucia-University of Pablo de Olavide-University of Seville-CSIC, Seville, Spain
| | - Petra I. Lorenzo
- Andalusian Center of Molecular Biology and Regenerative Medicine CABIMER, Junta de Andalucia-University of Pablo de Olavide-University of Seville-CSIC, Seville, Spain
| | - Benoit R. Gauthier
- Andalusian Center of Molecular Biology and Regenerative Medicine CABIMER, Junta de Andalucia-University of Pablo de Olavide-University of Seville-CSIC, Seville, Spain
- Centro de Investigacion Biomedica en Red de Diabetes y Enfermedades Metabolicas Asociadas (CIBERDEM), Madrid, Spain
| |
Collapse
|
9
|
Guo Y, Liu L, Cheng Y, Li H, Wan X, Ma J, Liu J, Liang W, Zhang P, Chen J, Cao X, Guan H, Xiao H, Li Y. Steroidogenic factor 1 protects mice from obesity-induced glucose intolerance via improving glucose-stimulated insulin secretion by beta cells. iScience 2023; 26:106451. [PMID: 37020955 PMCID: PMC10068556 DOI: 10.1016/j.isci.2023.106451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 02/12/2023] [Accepted: 03/15/2023] [Indexed: 04/07/2023] Open
Abstract
As a potential druggable nuclear receptor, steroidogenic factor 1 (SF1) regulates obesity and insulin resistance in the ventromedial hypothalamic nucleus. Herein, we sought to demonstrate its expression and functions in islets in the development of obesity-induced diabetes. SF1 was barely detected in the beta cells of lean mice but highly expressed in those of non-diabetic obese mice, while decreased in diabetic ones. Conditional deletion of SF1 in beta cells predisposed diet-induced obese (DIO) mice to glucose intolerance by perturbing glucose-stimulated insulin secretion (GSIS). Consistently, forced expression of SF1 restored favorable glucose homeostasis in DIO and db/db mice by improving GSIS. In isolated islets and MIN6, overexpression of SF1 also potentiated GSIS, mediated by improvement of mitochondrial ATP production. The underlying mechanisms may involve oxidative phosphorylation and lipid metabolism. Collectively, SF1 in beta cell preserves GSIS to promote beta-cell adaptation to obesity and hence is a potential therapeutic target for obesity-induced diabetes.
Collapse
Affiliation(s)
- Yan Guo
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Liehua Liu
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Yanglei Cheng
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Hai Li
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Xuesi Wan
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Jiajing Ma
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Juan Liu
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Weiwei Liang
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Pengyuan Zhang
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Jie Chen
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Xiaopei Cao
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Hongyu Guan
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
- Corresponding author
| | - Haipeng Xiao
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
- Corresponding author
| | - Yanbing Li
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
- Corresponding author
| |
Collapse
|
10
|
Meng L, Dong F, Deng J. NR5A2 as a potential target for exercise to improve metabolic syndrome. Aging (Albany NY) 2023; 15:2485-2502. [PMID: 37053002 PMCID: PMC10120892 DOI: 10.18632/aging.204606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/04/2023] [Indexed: 04/14/2023]
Abstract
BACKGROUND Metabolic syndrome is a syndrome of a variety of metabolic disorders. Exercise is beneficial to the human body. However, the association of NR5A2 and exercise with metabolic syndrome remains unclear. METHODS Download the GSE10540 and GSE12385 from GEO database. Bioinformatics analysis was used to screen the hub molecular of the metabolic syndrome. Forty 3-week-old C57BL/6J male mice were used in this study. The mean body weight was (17.5 ± 2.1) g. After 10 days of adaptive feeding, they were randomly divided into 4 groups according to the random number table method: Model + Exercise (n = 10), Model (n = 10), Model/NR5A2-OE (n = 10), Model/NR5A2-KO (n = 10). Western Blotting was performed to detect the expression of hub genes and signaling pathway. RESULTS There were 349 DEGs in GSE10540 and 49 DEGs in GSE12385. 10 core genes were obtained. GO showed that differentially expressed genes were mainly enriched in vascular morphogenesis, contractile fiber fraction, chemotaxis, and MAPK cascade regulation. KEGG showed that MAPK signaling pathway was a significant section in the metabolic syndrome. PIK3R2, STRA8, FLT1, DMRT1, FGF22, NR5A2, and FLT were up-regulated and PRDM14, POU5F1, and KDR were down-regulated in metabolic syndrome after exercise. CONCLUSION The expression of NR5A2 is down-regulated in metabolic syndrome, and exercise can increase the expression level of NR5A2. NR5A2 might be used as a potential target for exercise to improve metabolic syndrome.
Collapse
Affiliation(s)
- Lingxiu Meng
- Department of Cardiology, Qinhuangdao Second Hospital, Qinhuangdao, Hebei 066600, PR China
| | - Fusheng Dong
- Department of Anesthesiology, Qinhuangdao Second Hospital, Qinhuangdao, Hebei 066600, PR China
| | - Junguo Deng
- Department of Cardiology, Qinhuangdao Second Hospital, Qinhuangdao, Hebei 066600, PR China
| |
Collapse
|