1
|
Cazzaniga G, Capelli D, Montanari R, Fassi EMA, Grazioso G, Tresoldi A, Rinaldi F, Calleri E, Bassanini I, Romeo S, Garofalo M, Mori M, Meneghetti F, Villa S. Enhancing the activity of γ-hydroxy lactone derivatives as innovative peroxisome proliferator-activated receptor γ non-agonists inhibiting cyclin-dependent kinase 5-mediated phosphorylation. Eur J Med Chem 2025; 292:117657. [PMID: 40318479 DOI: 10.1016/j.ejmech.2025.117657] [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: 02/25/2025] [Revised: 04/09/2025] [Accepted: 04/17/2025] [Indexed: 05/07/2025]
Abstract
Insulin resistance (IR) is a pathological condition in which tissues exhibit a reduced response to normal or elevated levels of insulin. Type 2 diabetes mellitus (T2DM) and Metabolic Syndrome are the most prevalent disorders associated with IR. Most of the glitazones, traditional anti-diabetic drugs acting as Peroxisome Proliferator-Activated Receptor γ (PPARγ) agonists, have been withdrawn from the market. To mitigate the serious adverse effects associated with PPARγ agonism, a new opportunity is represented by the inhibitors of PPARγ phosphorylation by the Cyclin-Dependent Kinase 5 (CDK5). Their mechanism of action is mediated by the stabilization of the PPARγ β-sheet containing Ser245. Recently, we identified 4-(4-bromophenyl)-3-hydroxy-5-(3-hydroxyphenyl)furan-2(5H)-one (I) as a PPARγ non-agonist, capable of blocking the phosphorylation of the enzyme without direct effects on either CDK5 or PPARγ. Here, we isolated the two enantiomers of I, unambiguously defined their absolute configuration through single crystal X-ray diffraction and demonstrated by Grating-Coupled Interferometry binding assays that both (S)-I and (R)-I exhibited comparable affinity for PPARγ. Then, a library of 12 analogs was designed through structure-based modifications, optimizing the interactions within the ligand-binding domain. GCI analysis identified derivative 11, featuring an oxyacetic group in place of the initial hydroxyl function of the reference compound I, as the most promising candidate (KD = 186 nM). The crystal structure of the PPARγ-LBD/11 complex revealed a hydrogen bond interaction with Arg280, further stabilizing the binding conformation. These findings highlight the potential of γ-hydroxy lactone derivatives as PPARγ modulators and provide a foundation for future drug development targeting IR.
Collapse
Affiliation(s)
- Giulia Cazzaniga
- Department of Pharmaceutical Sciences, University of Milan, via L. Mangiagalli 25, 20133, Milano, Italy; Department of Science and High Technology, University of Insubria, via Valleggio 9, 22100, Como, Italy
| | - Davide Capelli
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche, Strada Provinciale 35d, n. 9-00010, Montelibretti, 34149, Rome, Italy
| | - Roberta Montanari
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche, Strada Provinciale 35d, n. 9-00010, Montelibretti, 34149, Rome, Italy
| | | | - Giovanni Grazioso
- Department of Pharmaceutical Sciences, University of Milan, via L. Mangiagalli 25, 20133, Milano, Italy
| | - Andrea Tresoldi
- Department of Pharmaceutical Sciences, University of Milan, via L. Mangiagalli 25, 20133, Milano, Italy
| | - Francesca Rinaldi
- Department of Drug Sciences, University of Pavia, via T. Taramelli 12, 27100, Pavia, Italy
| | - Enrica Calleri
- Department of Drug Sciences, University of Pavia, via T. Taramelli 12, 27100, Pavia, Italy
| | - Ivan Bassanini
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", Consiglio Nazionale delle Ricerche, Via Mario Bianco 9, 20131, Milano, Italy
| | - Sergio Romeo
- Department of Pharmaceutical Sciences, University of Milan, via L. Mangiagalli 25, 20133, Milano, Italy; Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", Consiglio Nazionale delle Ricerche, Via Mario Bianco 9, 20131, Milano, Italy
| | - Mariangela Garofalo
- Department of Pharmaceutical Sciences, University of Padova, via F. Marzolo 5, 35131, Padova, Italy
| | - Matteo Mori
- Department of Pharmaceutical Sciences, University of Milan, via L. Mangiagalli 25, 20133, Milano, Italy.
| | - Fiorella Meneghetti
- Department of Pharmaceutical Sciences, University of Milan, via L. Mangiagalli 25, 20133, Milano, Italy
| | - Stefania Villa
- Department of Pharmaceutical Sciences, University of Milan, via L. Mangiagalli 25, 20133, Milano, Italy
| |
Collapse
|
2
|
Han Z, Dong Q, Lu X, Liu S, Yang Y, Shao F, Tian L. TSH upregulates CYP4B1 through the PI3K/AKT/CREB pathway to promote cardiac hypertrophy. J Endocrinol Invest 2025:10.1007/s40618-025-02554-z. [PMID: 40056338 DOI: 10.1007/s40618-025-02554-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 02/09/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND Subclinical hypothyroidism (SCH) is closely associated with heart failure and cardiac hypertrophy, yet the underlying mechanism remains unclear. METHODS Cardiomyocytes treated with thyroid-stimulating hormone (TSH) were used as an in vitro model. Cardiac-specific TSHR knockout mice (CKO) were treated with isoproterenol (ISO) to induce cardiac hypertrophy in vivo. Serum FT4, TSH levels, heart weight, body weight and tibial length of mice were evaluated. Heart function was analyzed by M-mode cardiac ultrasonography. The pathological changes in cardiac tissues were detected by immunohistochemistry, hematoxylin-eosin and WGA staining. mRNA levels of ANP, BNP, α-MHC and β-MHC were evaluated by RT-PCR. Western blot was used to detect pathway related proteins. Besides, the transcriptome sequencing analysis and dual-luciferase reporter assays were used to verify the relevant molecular mechanisms. RESULTS TSH significantly promotes cardiomyocyte hypertrophy in cardiomyocytes. Meanwhile, cardiac-specific TSHR knockout significantly reduced ISO-induced cardiac hypertrophy. This was demonstrated by reductions in cell sizes, decreased HW/BW and HW/TL ratios, along with improved expression of hypertrophic genes. Further transcriptome sequencing results showed that TSH can significantly promote the expression of CYP4B1 in vitro. And the knockdown of CYP4B1 repressed TSH-induced cardiomyocyte hypertrophy. Further mechanistic studies revealed that TSH regulated the expression of CYP4B1 hypertrophy through the PI3K/AKT/CREB signaling pathway. Subsequently, the dual-luciferase assays demonstrated that CREB promotes the transcription of CYP4B1 by binding to its promoter region. CONCLUSION Overall, our findings highlight the direct impact of TSH/TSHR on cardiomyocyte hypertrophy and proposed CYP4B1 as a promising target for mitigating cardiac hypertrophy in SCH patients.
Collapse
Affiliation(s)
- Ziqi Han
- Gansu University of Chinese Medicine, Lanzhou, 730000, China
- Department of Endocrinology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Qianqian Dong
- Department of Endocrinology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Xiao Lu
- Gansu University of Chinese Medicine, Lanzhou, 730000, China
- Department of Endocrinology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Shanshan Liu
- Gansu University of Chinese Medicine, Lanzhou, 730000, China
- Department of Endocrinology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Yanlong Yang
- Gansu University of Chinese Medicine, Lanzhou, 730000, China
- Department of Endocrinology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Feifei Shao
- Department of Endocrinology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Limin Tian
- Gansu University of Chinese Medicine, Lanzhou, 730000, China.
- Department of Endocrinology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610000, China.
| |
Collapse
|
3
|
Yu H, Liu C, Zhang Y, Li Z, Pi W, Hu G. Regulation of PPARγ in the development of early sheep embryos in vitro. Theriogenology 2025; 234:143-150. [PMID: 39700755 DOI: 10.1016/j.theriogenology.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024]
Abstract
Lipid metabolism plays an important role in the regulation of early embryonic development in mammals. However, the effect of lipid metabolism mediated by peroxisome proliferator-activated receptor γ (PPARγ) on the early embryonic development of sheep remains unclear. In this study, rosiglitazone (RSG), a PPARγ activator, was added to the in vitro embryo culture (IVC) medium to regulate the continuous expression of PPARγ. This study aimed to evaluate PPARγ expression during early embryonic development in sheep as well as its effects on lipid deposition, reactive oxygen species (ROS) and glutathione (GSH) levels, apoptosis and lipid metabolism-related gene expression, and embryonic development. PPARγ was not detected at 2-cell, 4-cell, 8-cell, and morula stage, while widely expressed with obvious nuclear expression features in blastocysts. Notably, treatment with 5 μM RSG in sheep parthenogenetic activated (PA) embryos significantly increased the blastocyst rate, lipid content, and GSH levels, while decreasing ROS levels. Further analysis revealed that RSG treatment upregulated the expression levels of antioxidant genes (SOD2 and CAT), anti-apoptotic gene (BCL2), and lipid metabolism-related genes (SCD-1, CD36, PLIN2, FABP3, and FABP4). Taken together, these results suggest that PPARγ plays a vital role in promoting embryonic development by enhancing lipid metabolism and reducing oxidative stress.
Collapse
Affiliation(s)
- Hengbin Yu
- College of Animal Science and Technology, Shihezi University, Shihezi, 832003, China
| | - Chang Liu
- College of Animal Science and Technology, Shihezi University, Shihezi, 832003, China
| | - Yue Zhang
- College of Animal Science and Technology, Shihezi University, Shihezi, 832003, China
| | - Zhenghang Li
- College of Animal Science and Technology, Shihezi University, Shihezi, 832003, China
| | - Wenhui Pi
- College of Animal Science and Technology, Shihezi University, Shihezi, 832003, China.
| | - Guangdong Hu
- College of Animal Science and Technology, Shihezi University, Shihezi, 832003, China.
| |
Collapse
|
4
|
Długosz A, Błaszak B, Czarnecki D, Szulc J. Mechanism of Action and Therapeutic Potential of Xanthohumol in Prevention of Selected Neurodegenerative Diseases. Molecules 2025; 30:694. [PMID: 39942798 PMCID: PMC11821245 DOI: 10.3390/molecules30030694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/22/2025] [Accepted: 01/26/2025] [Indexed: 02/16/2025] Open
Abstract
Xanthohumol (XN), a bioactive plant flavonoid, is an antioxidant, and as such, it exhibits numerous beneficial properties, including anti-inflammatory, antimicrobial, and antioxidative effects. The main dietary source of XN is beer, where it is introduced through hops. Although the concentration of XN in beer is low, the large quantities of hop-related post-production waste present an opportunity to extract XN residues for technological or pharmaceutical purposes. The presented study focuses on the role of XN in the prevention of neurodegenerative diseases, analyzing its effect at a molecular level and including its signal transduction and metabolism. The paper brings up XN's mechanism of action, potential effects, and experimental and clinical studies on Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). Additionally, challenges and future research directions on XN, including its bioavailability, safety, and tolerance, have been discussed.
Collapse
Affiliation(s)
- Anna Długosz
- Faculty of Chemical Technology and Engineering, Department of Food Industry Technology and Engineering, Bydgoszcz University of Science and Technology, 85-326 Bydgoszcz, Poland;
| | - Błażej Błaszak
- Faculty of Chemical Technology and Engineering, Department of Food Industry Technology and Engineering, Bydgoszcz University of Science and Technology, 85-326 Bydgoszcz, Poland;
| | - Damian Czarnecki
- Faculty of Health Sciences, Department of Preventive Nursing, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-821 Bydgoszcz, Poland;
| | - Joanna Szulc
- Faculty of Chemical Technology and Engineering, Department of Food Industry Technology and Engineering, Bydgoszcz University of Science and Technology, 85-326 Bydgoszcz, Poland;
| |
Collapse
|
5
|
Chen F, Ma L, Liu Q, Zhou Z, Yi W. Recent advances and therapeutic applications of PPARγ-targeted ligands based on the inhibition mechanism of Ser273 phosphorylation. Metabolism 2025; 163:156097. [PMID: 39637972 DOI: 10.1016/j.metabol.2024.156097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/27/2024] [Accepted: 11/30/2024] [Indexed: 12/07/2024]
Abstract
PPARγ functions as a master ligand-dependent transcription factor that regulates the expressions of a variety of key genes related to metabolic homeostasis and inflammatory immunity. It has been recognized as a popular and druggable target in modern drug discovery. Similar to other nuclear receptors, PPARγ is a phosphoprotein, and its biological functions are regulated by phosphorylation, especially at Ser273 site which is mediated by CDK5 or ERK. In the past decade, the excessive level of PPARγ-Ser273 phosphorylation has been confirmed to be a crucial factor in promoting the occurrence and development of some major diseases. Ligands capable of inhibiting PPARγ-Ser273 phosphorylation have shown great potentials for treatment. Despite these achievements, to our knowledge, no related review focusing on this topic has been conducted so far. Therefore, we herein summarize the basic knowledge of PPARγ and CDK5/ERK-mediated PPARγ-Ser273 phosphorylation as well as its physiopathological role in representative diseases. We also review the developments and therapeutic applications of PPARγ-targeted ligands based on this mechanism. Finally, we suggest several directions for future investigations. We expect that this review can evoke more inspiration of scientific communities, ultimately facilitating the promotion of the PPARγ-Ser273 phosphorylation-involved mechanism as a promising breakthrough point for addressing the clinical treatment of human diseases.
Collapse
Affiliation(s)
- Fangyuan Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation & Molecular Target and Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Lei Ma
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation & Molecular Target and Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Qingmei Liu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation & Molecular Target and Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Zhi Zhou
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation & Molecular Target and Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China.
| | - Wei Yi
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation & Molecular Target and Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China.
| |
Collapse
|
6
|
Kononova YA, Tuchina TP, Babenko AY. Brown and Beige Adipose Tissue: One or Different Targets for Treatment of Obesity and Obesity-Related Metabolic Disorders? Int J Mol Sci 2024; 25:13295. [PMID: 39769065 PMCID: PMC11677471 DOI: 10.3390/ijms252413295] [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: 10/06/2024] [Revised: 11/27/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
The failure of the fight against obesity makes us turn to new goals in its treatment. Now, brown adipose tissue has attracted attention as a promising target for the treatment of obesity and associated metabolic disorders such as insulin resistance, dyslipidemia, and glucose tolerance disorders. Meanwhile, the expansion of our knowledge has led to awareness about two rather different subtypes: classic brown and beige (inducible brown) adipose tissue. These subtypes have different origin, differences in the expression of individual genes but also a lot in common. Both tissues are thermogenic, which means that, by increasing energy consumption, they can improve their balance with excess intake. Both tissues are activated in response to specific inducers (cold, beta-adrenergic receptor activation, certain food and drugs), but beige adipose tissue transdifferentiates back into white adipose tissue after the cessation of inducing action, while classic brown adipose tissue persists, but its activity decreases. In this review, we attempted to understand whether there are differences in the effects of different groups of thermogenesis-affecting drugs on these tissues. The analysis showed that this area of research is rather sparse and requires close attention in further studies.
Collapse
Affiliation(s)
- Yulia A. Kononova
- World-Class Scientific Center “Center for Personalized Medicine”, Almazov National Medical Research Centre, 197341 St. Petersburg, Russia;
| | - Taisiia P. Tuchina
- Endocrinology Department, Almazov National Medical Research Centre, 197341 St. Petersburg, Russia;
| | - Alina Yu. Babenko
- World-Class Scientific Center “Center for Personalized Medicine”, Almazov National Medical Research Centre, 197341 St. Petersburg, Russia;
| |
Collapse
|
7
|
Xue P, You X, Ren L, Yue W, Ma Z. PPARγ-mediated amelioration of lipid metabolism abnormality by kaempferol. Arch Biochem Biophys 2024; 761:110154. [PMID: 39278305 DOI: 10.1016/j.abb.2024.110154] [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: 04/25/2024] [Revised: 09/04/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024]
Abstract
Kaempferol can exert biological functions by regulating various signaling pathways. This study evaluated the ameliorative effect of kaempferol on lipid accumulation using oleic acid and palmitic acid-treated HepG2 cells and high-fat diet mice. In vitro oil red O staining showed that kaempferol treatment improved lipid accumulation (p < 0.001 for TG content and p < 0.05 for TC content). Immunofluorescence, Western blot analysis and RT-qPCR showed that kaempferol could promote nuclear translocation of PPARγ and reduce the expression of PPARγ, C/EBPβ, and SREBP-1c. Dietary intervention with kaempferol could reduce the lipid accumulation in hepatocytes and inflammatory cell infiltration, as well as attenuated serum levels of IL-6 and TNF-α in HFD-fed mice (p < 0.001 for IL-6 and p < 0.01 for TNF-α at kaempferol 60 mg/kg/d). Meanwhile, histopathological examination revealed that there was no substantial damage or distinct inflammation lesions in organs at the experimental dose, including the heart, lung, kidney, and spleen. The aforementioned research findings can serve as references for further preclinical investigations on the potential of kaempferol to mitigate lipid accumulation.
Collapse
Affiliation(s)
- Peiyu Xue
- School of Biology and Food Engineering, Anyang Institute of Technology, Anyang, 455000, China
| | - Xinyong You
- School of Biology and Food Engineering, Anyang Institute of Technology, Anyang, 455000, China
| | - Li Ren
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China
| | - Weiming Yue
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, China.
| | - Zheng Ma
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, China.
| |
Collapse
|
8
|
Boretto C, Muzio G, Autelli R. PPARγ antagonism as a new tool for preventing or overcoming endocrine resistance in luminal A breast cancers. Biomed Pharmacother 2024; 180:117461. [PMID: 39326102 DOI: 10.1016/j.biopha.2024.117461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/10/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024] Open
Abstract
PURPOSE This research investigates the role of PPARγ in the complex molecular events underlying the acquisition of resistance to tamoxifen (Tam) in luminal A breast cancer (BC) cells. Furthermore, it focuses on evaluating the possibility of repurposing Imatinib mesylate, an FDA-approved anticancer agent recently recognized also as a PPARγ antagonist, for the personalized therapy of endocrine-resistant BC with increased PPARγ expression. METHODS Differential gene expression between parental and Tam-resistant MCF7 cells was assessed by RNA-seq followed by bioinformatics analysis and validation by RT-qPCR. PPARγ was downregulated by esiRNAs or inhibited by the antagonist GW9662. Cell viability and proliferation were measured by MTT and colony formation assays. Spheroids were prepared from parental and Tam-resistant MCF7 cells. Other luminal A BC cell lines resistant to Tam were generated. RESULTS In MCF7-TamR cells, PPARγ and several of its target genes were significantly upregulated. Increased PPARγ expression was due to the modulation of its positive/negative transcriptional regulators. Downregulating PPARγ with esiRNAs or GW9662 effectively killed parental and Tam-resistant cells and spheroids. Imatinib revealed to be as effective as GW9662 in restoring Tam susceptibility of these cells. PPARγ overexpression was also observed in the newly-selected Tam-resistant luminal A BC cells, in which GW9662 and Imatinib restored their susceptibility to Tam. CONCLUSION Our findings demonstrate that the overexpression of PPARγ is a frequent occurrence during acquisition of Tam resistance in luminal A BC cells, and that PPARγ antagonism represents an alternative therapeutic approach for the personalized treatment of BC showing dysregulation of this nuclear receptor.
Collapse
Affiliation(s)
- Cecilia Boretto
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, Turin 10125, Italy
| | - Giuliana Muzio
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, Turin 10125, Italy
| | - Riccardo Autelli
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, Turin 10125, Italy.
| |
Collapse
|
9
|
Ma L, Tang J, Chen F, Liu Q, Huang J, Liu X, Zhou Z, Yi W. Structure-based screening, optimization and biological evaluation of novel chrysin-based derivatives as selective PPARγ modulators for the treatment of T2DM and hepatic steatosis. Eur J Med Chem 2024; 276:116728. [PMID: 39089002 DOI: 10.1016/j.ejmech.2024.116728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/24/2024] [Accepted: 07/28/2024] [Indexed: 08/03/2024]
Abstract
In consideration of several serious side effects induced by the classical AF-2 involved "lock" mechanism, recently disclosed PPARγ-Ser273 phosphorylation mode of action has become an alternative and mainstream mechanism for currently PPARγ-based drug discovery and development with an improved therapeutic index. In this study, by virtue of structure-based virtual high throughput screening (SB-VHTS), structurally chemical optimization by targeting the inhibition of the PPARγ-Ser273 phosphorylation as well as in vitro biological evaluation, which led to the final identification of a chrysin-based potential hit (YGT-31) as a novel selective PPARγ modulator with potent binding affinity and partial agonism. Further in vivo evaluation demonstrated that YGT-31 possessed potent glucose-lowering and relieved hepatic steatosis effects without involving the TZD-associated side effects. Mechanistically, YGT-31 presented such desired therapeutic index, mainly because it effectively inhibited the CDK5-mediated PPARγ-Ser273 phosphorylation, selectively elevated the level of insulin sensitivity-related Glut4 and adiponectin but decreased the expression of insulin-resistance-associated genes PTP1B and SOCS3 as well as inflammation-linked genes IL-6, IL-1β and TNFα. Finally, the molecular docking study was also conducted to uncover an interesting hydrogen-bonding network of YGT-31 with PPARγ-Ser273 phosphorylation-related key residues Ser342 and Glu343, which not only gave a clear verification for our targeting modification but also provided a proof of concept for the abovementioned molecular mechanism.
Collapse
Affiliation(s)
- Lei Ma
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Junyuan Tang
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China; Department of Food and Chemical Engineering, Shaoyang University, Shao Shui Xi Road, Shaoyang, 422100, China
| | - Fangyuan Chen
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Qingmei Liu
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Junjun Huang
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Xiawen Liu
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China.
| | - Zhi Zhou
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China.
| | - Wei Yi
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China.
| |
Collapse
|
10
|
Ma Y, Lai J, Chen Z, Wan Q, Shi X, Zhou H, Li J, Yang Z, Wu J. Exploring therapeutic targets and molecular mechanisms for treating diabetes mellitus-associated heart failure with Qishen Yiqi dropping pills: A network pharmacology and bioinformatics approach. Medicine (Baltimore) 2024; 103:e39104. [PMID: 39093800 PMCID: PMC11296435 DOI: 10.1097/md.0000000000039104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/01/2024] [Indexed: 08/04/2024] Open
Abstract
Diabetes mellitus (DM) and heart failure frequently coexist, presenting significant public health challenges. QiShenYiQi Dropping Pills (QSDP) are widely employed in the treatment of diabetes mellitus concomitant with heart failure (DM-HF). Nevertheless, the precise mechanisms underlying their efficacy have yet to be elucidated. Active ingredients and likely targets of QSDP were retrieved from the TCMSP and UniProt databases. Genes associated with DM-HF were pinpointed through searches in the GeneCards, OMIM, DisGeNET, and TTD databases. Differential genes connected to DM-HF were sourced from the GEO database. Enrichment analyses via gene ontology and Kyoto Encyclopedia of Genes and Genomes pathways, as well as immune infiltration assessments, were conducted using R software. Further analysis involved employing molecular docking strategies to explore the interactions between the identified targets and active substances in QSDP that are pertinent to DM-HF treatment. This investigation effectively discerned 108 active compounds and 257 targets relevant to QSDP. A protein-protein interaction network was constructed, highlighting 6 central targets for DM-HF treatment via QSDP. Gene ontology enrichment analysis predominantly linked these targets with responses to hypoxia, metabolism of reactive oxygen species, and cytokine receptor interactions. Analysis of Kyoto Encyclopedia of Genes and Genomes pathways demonstrated that these targets mainly participate in pathways linked to diabetic complications, such as AGE-RAGE signaling, dyslipidemia, arteriosclerosis, the HIF-1 signaling pathway, and the tumor necrosis factor signaling pathway. Further, immune infiltration analysis implied that QSDP's mechanism in treating DM-HF might involve immune-mediated inflammation and crucial signaling pathways. Additionally, molecular docking studies showed that the active substances in QSDP have strong binding affinities with these identified targets. This research presents a new model for addressing DM-HF through the use of QSDP, providing novel insights into incorporating traditional Chinese medicine (TCM) principles in the clinical treatment of DM-HF. The implications of these findings are substantial for both clinical application and further scientific inquiry.
Collapse
Affiliation(s)
- Yirong Ma
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Junyu Lai
- Cardiology Department, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Zhengtao Chen
- Cardiology Department, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Qiang Wan
- Cardiology Department, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Xianlin Shi
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Hao Zhou
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Jiaming Li
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Zurong Yang
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Jianguang Wu
- Cardiology Department, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| |
Collapse
|
11
|
Chu Y, Gui S, Zheng Y, Zhao J, Zhao Y, Li Y, Chen X. The natural compounds, Magnolol or Honokiol, promote adipose tissue browning and resist obesity through modulating PPARα/γ activity. Eur J Pharmacol 2024; 969:176438. [PMID: 38402928 DOI: 10.1016/j.ejphar.2024.176438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 02/11/2024] [Accepted: 02/16/2024] [Indexed: 02/27/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is closely associated with the body's energy metabolism. A potential strategy to regulate energy metabolism, combat obesity, and reduce NAFLD is by enhancing adipocyte thermogenesis and increasing energy expenditure. In this study, our objective was to examine the effects of phenolic extracts derived from Magnolia officinalis on the regulation of NAFLD. Specifically, we investigated the impact of Magnolol or Honokiol treatment on high-fat diet (HFD)-induced obese C57BL6/J male mice. Firstly, we monitored energy metabolism, dissected tissues, and analyzed tissue sections. Additionally, we conducted experiments on HepG2 and primary adipocytes to gain insights into the roles of Magnolol or Honokiol. To further understand the effects of these compounds on related signaling pathways and marker genes, we performed molecular docking, dual-luciferase assays, and interfered with target genes. Our findings revealed that Magnolol or Honokiol activate the peroxisome proliferator activated receptor alpha (PPARα) signaling pathway, leading to the alleviation of NAFLD. This activation promotes fatty acid oxidation, reduces lipogenesis, and enhances the expression and secretion of FGF21. Notably, Fibroblast growth factor 21 (FGF21), secreted by the liver, plays a crucial role in improving communication between the liver and adipocytes while also promoting the browning of adipose tissue. Additionally, Magnolol or Honokiol activate the peroxisome proliferator activated receptor gamma (PPARγ) signaling pathway, resulting in increased uncoupling protein 1 (UCP1) expression, heightened heat production in adipose tissue, and anti-obesity. Therefore, Magnolol or Honokiol alleviate NAFLD, promote adipose tissue browning and resist obesity through dual activation of PPARα/γ.
Collapse
Affiliation(s)
- Yi Chu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology &College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Sisi Gui
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology &College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yazhen Zheng
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology &College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jingwu Zhao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology &College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yaxiang Zhao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology &College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yingying Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology &College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xiaodong Chen
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology &College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
12
|
Liu Q, Ma L, Chen F, Zhang S, Huang Z, Zheng X, Chen Z, Ye J, Hou N, Yi W, Zhou Z. Raloxifene-driven benzothiophene derivatives: Discovery, structural refinement, and biological evaluation as potent PPARγ modulators based on drug repurposing. Eur J Med Chem 2024; 269:116325. [PMID: 38527378 DOI: 10.1016/j.ejmech.2024.116325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/26/2024] [Accepted: 03/09/2024] [Indexed: 03/27/2024]
Abstract
By virtue of the drug repurposing strategy, the anti-osteoporosis drug raloxifene was identified as a novel PPARγ ligand through structure-based virtual high throughput screening (SB-VHTS) of FDA-approved drugs and TR-FRET competitive binding assay. Subsequent structural refinement of raloxifene led to the synthesis of a benzothiophene derivative, YGL-12. This compound exhibited potent PPARγ modulation with partial agonism, uniquely promoting adiponectin expression and inhibiting PPARγ Ser273 phosphorylation by CDK5 without inducing the expression of adipongenesis associated genes, including PPARγ, aP2, CD36, FASN and C/EBPα. This specific activity profile resulted in effective hypoglycemic properties, avoiding major TZD-related adverse effects like weight gain and hepatomegaly, which were demonstrated in db/db mice. Molecular docking studies showed that YGL-12 established additional hydrogen bonds with Ile281 and enhanced hydrogen-bond interaction with Ser289 as well as PPARγ Ser273 phosphorylation-related residues Ser342 and Glu343. These findings suggested YGL-12 as a promising T2DM therapeutic candidate, thereby providing a molecular framework for the development of novel PPARγ modulators with an enhanced therapeutic index.
Collapse
Affiliation(s)
- Qingmei Liu
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Lei Ma
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Fangyuan Chen
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Shuyun Zhang
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Zexin Huang
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Xiufen Zheng
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Zikai Chen
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Junwei Ye
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Ning Hou
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China.
| | - Wei Yi
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China.
| | - Zhi Zhou
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China.
| |
Collapse
|
13
|
Ghesmati Z, Rashid M, Fayezi S, Gieseler F, Alizadeh E, Darabi M. An update on the secretory functions of brown, white, and beige adipose tissue: Towards therapeutic applications. Rev Endocr Metab Disord 2024; 25:279-308. [PMID: 38051471 PMCID: PMC10942928 DOI: 10.1007/s11154-023-09850-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/30/2023] [Indexed: 12/07/2023]
Abstract
Adipose tissue, including white adipose tissue (WAT), brown adipose tissue (BAT), and beige adipose tissue, is vital in modulating whole-body energy metabolism. While WAT primarily stores energy, BAT dissipates energy as heat for thermoregulation. Beige adipose tissue is a hybrid form of adipose tissue that shares characteristics with WAT and BAT. Dysregulation of adipose tissue metabolism is linked to various disorders, including obesity, type 2 diabetes, cardiovascular diseases, cancer, and infertility. Both brown and beige adipocytes secrete multiple molecules, such as batokines, packaged in extracellular vesicles or as soluble signaling molecules that play autocrine, paracrine, and endocrine roles. A greater understanding of the adipocyte secretome is essential for identifying novel molecular targets in treating metabolic disorders. Additionally, microRNAs show crucial roles in regulating adipose tissue differentiation and function, highlighting their potential as biomarkers for metabolic disorders. The browning of WAT has emerged as a promising therapeutic approach in treating obesity and associated metabolic disorders. Many browning agents have been identified, and nanotechnology-based drug delivery systems have been developed to enhance their efficacy. This review scrutinizes the characteristics of and differences between white, brown, and beige adipose tissues, the molecular mechanisms involved in the development of the adipocytes, the significant roles of batokines, and regulatory microRNAs active in different adipose tissues. Finally, the potential of WAT browning in treating obesity and atherosclerosis, the relationship of BAT with cancer and fertility disorders, and the crosstalk between adipose tissue with circadian system and circadian disorders are also investigated.
Collapse
Affiliation(s)
- Zeinab Ghesmati
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohsen Rashid
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shabnam Fayezi
- Department of Gynecologic Endocrinology and Fertility Disorders, Women's Hospital, Ruprecht-Karls University of Heidelberg, Heidelberg, Germany
| | - Frank Gieseler
- Division of Experimental Oncology, Department of Hematology and Oncology, University Medical Center Schleswig-Holstein, Campus Lübeck, 23538, Lübeck, Germany
| | - Effat Alizadeh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Masoud Darabi
- Division of Experimental Oncology, Department of Hematology and Oncology, University Medical Center Schleswig-Holstein, Campus Lübeck, 23538, Lübeck, Germany.
| |
Collapse
|
14
|
McClave SA, Martindale RG. Browning of white adipose tissue may be an appropriate adaptive response to critical illness. JPEN J Parenter Enteral Nutr 2024; 48:37-45. [PMID: 37908064 DOI: 10.1002/jpen.2576] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/16/2023] [Accepted: 10/28/2023] [Indexed: 11/02/2023]
Abstract
Both the baseline amount of brown adipose tissue (BAT) and the capacity to stimulate browning of white adipose tissue (WAT) may provide a protective effect to the patient in a critical care setting. Critical illness is associated with reduced mitochondrial volume and function resulting in the increased production of reactive oxygen species, greater demand for adenosine triphosphate, a switch to uncoupled fat metabolism, and hibernation of the organelle, which all contribute to multiple organ failure. Increasing insulin resistance, decreasing fatty acid oxidation, and dependence on carbohydrate metabolism result. Browning of WAT may oppose many of these adverse effects. The presence of BAT and the changes associated with browning may help dissipate oxidative stress, increase consumption and utilization of metabolites, and reduce pro-inflammatory actions. The number of mitochondria increases, and there is greater infiltration of macrophages into adipose tissue. A shift occurs in macrophage expression from the M1 to M2 phenotype, an effect which further dampens inflammation, increases insulin sensitivity, and improves tissue healing and remodeling. Any benefit from these responses may be lost in the disease states of chronic hypermetabolism (such as burns or cancer cachexia) in which the persistence of these physiologic effects may become detrimental, contributing to excessive weight loss, adipose wasting, and loss of lean body mass. This paper discusses the plasticity of adipose tissue and whether shifts in its physiology provide clinical advantages in the intensive care unit.
Collapse
Affiliation(s)
- Stephen A McClave
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Robert G Martindale
- Department of Surgery, Oregon Health Sciences University, Portland, Oregon, USA
| |
Collapse
|
15
|
Watanabe S, Haruyama R, Umezawa K, Tomioka I, Nakamura S, Katayama S, Mitani T. Genistein enhances NAD + biosynthesis by upregulating nicotinamide phosphoribosyltransferase in adipocytes. J Nutr Biochem 2023; 121:109433. [PMID: 37648097 DOI: 10.1016/j.jnutbio.2023.109433] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 08/16/2023] [Accepted: 08/25/2023] [Indexed: 09/01/2023]
Abstract
A decrease in the NAD+ level in adipocytes causes adipose-tissue dysfunction, leading to systemic glucose, and lipid metabolism failure. Therefore, it is necessary to develop small molecules and nutraceuticals that can increase NAD+ levels in adipocytes. Genistein, a nutraceutical derived from soybeans, has various physiological activities and improves glucose and lipid metabolism. In this study, we aimed to unravel the effects of genistein on the NAD+ level in adipocytes and the underlying molecular mechanisms. Genistein enhanced NAD+ biosynthesis by increasing the expression of nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme in NAD+ biosynthesis. A pull-down assay using genistein-immobilized beads revealed prohibitin 1 (PHB1) as a target protein of genistein. The knockdown of Phb1 suppressed the genistein-induced increase in NAMPT expression and NAD+ level in adipocytes. Genistein-bound PHB1 contributed to the stabilization of the transcription factor CCAAT/enhancer-binding protein β through the activation of extracellular signal-regulated kinase, resulting in increased NAMPT expression at the transcriptional level. Genistein induced the dephosphorylation of peroxisome proliferator-activated receptor at serine 273 and increased the level of the insulin-sensitizing adipokine adiponectin in adipocytes, whereas the knockdown of Nampt and Phb1 abolished these genistein-mediated effects. Our results proved the potential efficacy of genistein in increasing the NAD+ level and restoring metabolic function in adipocytes. Furthermore, we identified PHB1, localized to the plasma membrane, as a novel candidate target protein for increased expression of NAMPT in adipocytes. Overall, these findings will assist in developing NAD+-boosting nutraceuticals to alleviate metabolic dysfunctions in adipose tissues.
Collapse
Affiliation(s)
- Shun Watanabe
- Division of Food Science and Biotechnology, Graduate School of Science and Technology, Shinshu University, Nagano, Japan
| | - Riki Haruyama
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, Nagano, Japan
| | - Koji Umezawa
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, Japan
| | - Ikuo Tomioka
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, Nagano, Japan; Division of Biotechnology, Graduate School of Science and Technology, Shinshu University, Nagano, Japan
| | - Soichiro Nakamura
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, Nagano, Japan
| | - Shigeru Katayama
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, Japan
| | - Takakazu Mitani
- Division of Food Science and Biotechnology, Graduate School of Science and Technology, Shinshu University, Nagano, Japan; Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, Nagano, Japan.
| |
Collapse
|
16
|
Sadeghi A, Niknam M, Momeni-Moghaddam MA, Shabani M, Aria H, Bastin A, Teimouri M, Meshkani R, Akbari H. Crosstalk between autophagy and insulin resistance: evidence from different tissues. Eur J Med Res 2023; 28:456. [PMID: 37876013 PMCID: PMC10599071 DOI: 10.1186/s40001-023-01424-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 10/03/2023] [Indexed: 10/26/2023] Open
Abstract
Insulin is a critical hormone that promotes energy storage in various tissues, as well as anabolic functions. Insulin resistance significantly reduces these responses, resulting in pathological conditions, such as obesity and type 2 diabetes mellitus (T2DM). The management of insulin resistance requires better knowledge of its pathophysiological mechanisms to prevent secondary complications, such as cardiovascular diseases (CVDs). Recent evidence regarding the etiological mechanisms behind insulin resistance emphasizes the role of energy imbalance and neurohormonal dysregulation, both of which are closely regulated by autophagy. Autophagy is a conserved process that maintains homeostasis in cells. Accordingly, autophagy abnormalities have been linked to a variety of metabolic disorders, including insulin resistance, T2DM, obesity, and CVDs. Thus, there may be a link between autophagy and insulin resistance. Therefore, the interaction between autophagy and insulin function will be examined in this review, particularly in insulin-responsive tissues, such as adipose tissue, liver, and skeletal muscle.
Collapse
Affiliation(s)
- Asie Sadeghi
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Niknam
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Maryam Shabani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Aria
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Alireza Bastin
- Clinical Research Development Center "The Persian Gulf Martyrs" Hospital, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Maryam Teimouri
- Department of Biochemistry, School of Allied Medical Sciences, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Reza Meshkani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Akbari
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran.
- Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
17
|
Kim YC, Ki SW, Kim H, Kang S, Kim H, Go GW. Recent Advances in Nutraceuticals for the Treatment of Sarcopenic Obesity. Nutrients 2023; 15:3854. [PMID: 37686886 PMCID: PMC10490319 DOI: 10.3390/nu15173854] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 08/26/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023] Open
Abstract
Sarcopenic obesity, low muscle mass, and high body fat are growing health concerns in the aging population. This review highlights the need for standardized criteria and explores nutraceuticals as potential therapeutic agents. Sarcopenic obesity is associated with insulin resistance, inflammation, hormonal changes, and reduced physical activity. These factors lead to impaired muscle activity, intramuscular fat accumulation, and reduced protein synthesis, resulting in muscle catabolism and increased fat mass. Myostatin and irisin are myokines that regulate muscle synthesis and energy expenditure, respectively. Nutritional supplementation with vitamin D and calcium is recommended for increasing muscle mass and reducing body fat content. Testosterone therapy decreases fat mass and improves muscle strength. Vitamin K, specifically menaquinone-4 (MK-4), improves mitochondrial function and reduces muscle damage. Irisin is a hormone secreted during exercise that enhances oxidative metabolism, prevents insulin resistance and obesity, and improves bone quality. Low-glycemic-index diets and green cardamom are potential methods for managing sarcopenic obesity. In conclusion, along with exercise and dietary support, nutraceuticals, such as vitamin D, calcium, vitamin K, and natural agonists of irisin or testosterone, can serve as promising future therapeutic alternatives.
Collapse
Affiliation(s)
| | | | | | | | | | - Gwang-woong Go
- Department of Food and Nutrition, Hanyang University, Seoul 04763, Republic of Korea; (Y.-C.K.); (S.-W.K.); (H.K.); (S.K.); (H.K.)
| |
Collapse
|
18
|
Yu L, Gao Y, Aaron N, Qiang L. A glimpse of the connection between PPARγ and macrophage. Front Pharmacol 2023; 14:1254317. [PMID: 37701041 PMCID: PMC10493289 DOI: 10.3389/fphar.2023.1254317] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 07/31/2023] [Indexed: 09/14/2023] Open
Abstract
Nuclear receptors are ligand-regulated transcription factors that regulate vast cellular activities and serve as an important class of drug targets. Among them, peroxisome proliferator-activated receptors (PPARs) are members of the nuclear receptor family and have been extensively studied for their roles in metabolism, differentiation, development, and cancer, among others. Recently, there has been considerable interest in understanding and defining the function of PPARs and their agonists in regulating innate and adaptive immune responses and their pharmacological potential in combating chronic inflammatory diseases. In this review, we focus on emerging evidence for the potential role of PPARγ in macrophage biology, which is the prior innate immune executive in metabolic and tissue homeostasis. We also discuss the role of PPARγ as a regulator of macrophage function in inflammatory diseases. Lastly, we discuss the possible application of PPARγ antagonists in metabolic pathologies.
Collapse
Affiliation(s)
- Lexiang Yu
- Naomi Berrie Diabetes Center, Columbia University, New York, NY, United States
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Yuen Gao
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| | - Nicole Aaron
- Naomi Berrie Diabetes Center, Columbia University, New York, NY, United States
- Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, NY, United States
| | - Li Qiang
- Naomi Berrie Diabetes Center, Columbia University, New York, NY, United States
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| |
Collapse
|
19
|
Capelli D, Cazzaniga G, Mori M, Laghezza A, Loiodice F, Quaglia M, Negro E, Meneghetti F, Villa S, Montanari R. Biological Screening and Crystallographic Studies of Hydroxy γ-Lactone Derivatives to Investigate PPARγ Phosphorylation Inhibition. Biomolecules 2023; 13:biom13040694. [PMID: 37189440 DOI: 10.3390/biom13040694] [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: 02/24/2023] [Revised: 03/31/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
PPARγ represents a key target for the treatment of type 2 diabetes and metabolic syndrome. To avoid serious adverse effects related to the PPARγ agonism profile of traditional antidiabetic drugs, a new opportunity is represented by the development of molecules acting as inhibitors of PPARγ phosphorylation by the cyclin-dependent kinase 5 (CDK5). Their mechanism of action is mediated by the stabilization of the PPARγ β-sheet containing Ser273 (Ser245 in PPARγ isoform 1 nomenclature). In this paper, we report the identification of new γ-hydroxy-lactone-based PPARγ binders from the screening of an in-house library. These compounds exhibit a non-agonist profile towards PPARγ, and one of them prevents Ser245 PPARγ phosphorylation by acting mainly on PPARγ stabilization and exerting a weak CDK5 inhibitory effect.
Collapse
Affiliation(s)
- Davide Capelli
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche, Strada Provinciale 35d, n. 9-00010, Montelibretti, 34149 Rome, Italy
| | - Giulia Cazzaniga
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milano, Italy
| | - Matteo Mori
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milano, Italy
| | - Antonio Laghezza
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", Via Orabona 4, 70125 Bari, Italy
| | - Fulvio Loiodice
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", Via Orabona 4, 70125 Bari, Italy
| | - Martina Quaglia
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milano, Italy
| | - Elisa Negro
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche, Strada Provinciale 35d, n. 9-00010, Montelibretti, 34149 Rome, Italy
| | - Fiorella Meneghetti
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milano, Italy
| | - Stefania Villa
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milano, Italy
| | - Roberta Montanari
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche, Strada Provinciale 35d, n. 9-00010, Montelibretti, 34149 Rome, Italy
| |
Collapse
|
20
|
Yu J, Hu Y, Sheng M, Gao M, Guo W, Zhang Z, Wang D, Wu X, Li J, Chen Y, Zhao W, Liu C, Cui X, Chen X, Zhao C, Chen H, Xiao J, Chen S, Luo C, Xu L, Gu X, Ma X. Selective PPARγ modulator diosmin improves insulin sensitivity and promotes browning of white fat. J Biol Chem 2023; 299:103059. [PMID: 36841479 PMCID: PMC10033317 DOI: 10.1016/j.jbc.2023.103059] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 02/17/2023] [Accepted: 02/18/2023] [Indexed: 02/27/2023] Open
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) is a master regulator of adipocyte differentiation, glucolipid metabolism, and inflammation. Thiazolidinediones are PPARγ full agonists with potent insulin-sensitizing effects, whereas their oral usage is restricted because of unwanted side effects, including obesity and cardiovascular risks. Here, via virtual screening, microscale thermophoresis analysis, and molecular confirmation, we demonstrate that diosmin, a natural compound of wide and long-term clinical use, is a selective PPARγ modulator that binds to PPARγ and blocks PPARγ phosphorylation with weak transcriptional activity. Local diosmin administration in subcutaneous fat (inguinal white adipose tissue [iWAT]) improved insulin sensitivity and attenuated obesity via enhancing browning of white fat and energy expenditure. Besides, diosmin ameliorated inflammation in WAT and liver and reduced hepatic steatosis. Of note, we determined that iWAT local administration of diosmin did not exhibit obvious side effects. Taken together, the present study demonstrated that iWAT local delivery of diosmin protected mice from diet-induced insulin resistance, obesity, and fatty liver by blocking PPARγ phosphorylation, without apparent side effects, making it a potential therapeutic agent for the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Jian Yu
- Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China; Joint Center for Translational Medicine, Fengxian District Central Hospital, Shanghai, China
| | - Yepeng Hu
- Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Maozheng Sheng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Mingyuan Gao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Wenxiu Guo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Zhe Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Dongmei Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Xia Wu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Jin Li
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Sciences, Shanghai University, Shanghai, China
| | - Yantao Chen
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, The Center for Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Wenjun Zhao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Caizhi Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Xiangdi Cui
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Xin Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Cheng Zhao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Huang Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Sciences, Shanghai University, Shanghai, China
| | - Shijie Chen
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, The Center for Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Cheng Luo
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, The Center for Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Lingyan Xu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China.
| | - Xuejiang Gu
- Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Xinran Ma
- Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China; Joint Center for Translational Medicine, Fengxian District Central Hospital, Shanghai, China; Chongqing Key Laboratory of Precision Optics, Chongqing Institute of East China Normal University, Chongqing, China.
| |
Collapse
|
21
|
Guru B, Tamrakar AK, Manjula S, Prashantha Kumar B. Novel dual PPARα/γ agonists protect against liver steatosis and improve insulin sensitivity while avoiding side effects. Eur J Pharmacol 2022; 935:175322. [PMID: 36228743 DOI: 10.1016/j.ejphar.2022.175322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/04/2022] [Accepted: 10/05/2022] [Indexed: 11/03/2022]
|
22
|
KITLG Promotes Glomerular Endothelial Cell Injury in Diabetic Nephropathy by an Autocrine Effect. Int J Mol Sci 2022; 23:ijms231911723. [PMID: 36233032 PMCID: PMC9569900 DOI: 10.3390/ijms231911723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/29/2022] [Accepted: 09/29/2022] [Indexed: 11/18/2022] Open
Abstract
Diabetic nephropathy (DN) is an increasing threat to human health. The impact of hyperglycemia or its metabolites, advanced glycation end-products (AGEs), on glomerular endothelial cells (GECs) and their pathophysiologic mechanisms are not well explored. Our results reveal that AGEs increased the expression and secretion of the KIT ligand (KITLG) in GECs. Both AGEs and KITLG promoted endothelial-to-mesenchymal transition (EndoMT) in GECs and further increased the permeability of GECs through the AKT/extracellular-signal-regulated kinase pathway. Inhibition of KITLG’s effects by imatinib prevented AGE-medicated EndoMT in GECs, supporting the belief that KITLG is a critical factor for GEC injury. We found higher KITLG levels in the GECs and urine of db/db mice compared with db/m mice, and urinary KITLG levels were positively correlated with the urinary albumin-to-creatinine ratio (ACR). Furthermore, type 2 diabetic patients had higher urinary KITLG levels than normal individuals, as well as urinary KITLG levels that were positively correlated with urinary ACR and negatively correlated with the estimated glomerular filtration rate. KITLG plays a pathogenic role in GEC injury in DN and might act as a biomarker of DN progression.
Collapse
|
23
|
Ahn D, Kim J, Nam G, Zhao X, Kwon J, Hwang JY, Kim JK, Yoon SY, Chung SJ. Ethyl Gallate Dual-Targeting PTPN6 and PPARγ Shows Anti-Diabetic and Anti-Obese Effects. Int J Mol Sci 2022; 23:ijms23095020. [PMID: 35563411 PMCID: PMC9105384 DOI: 10.3390/ijms23095020] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 02/04/2023] Open
Abstract
The emergence of the high correlation between type 2 diabetes and obesity with complicated conditions has led to the coinage of the term “diabesity”. AMP-activated protein kinase (AMPK) activators and peroxisome proliferator-activated receptor (PPARγ) antagonists have shown therapeutic activity for diabesity, respectively. Hence, the discovery of compounds that activate AMPK as well as antagonize PPARγ may lead to the discovery of novel therapeutic agents for diabesity. In this study, the knockdown of PTPN6 activated AMPK and suppressed adipogenesis in 3T3-L1 cells. By screening a library of 1033 natural products against PTPN6, we found ethyl gallate to be the most selective inhibitor of PTPN6 (Ki = 3.4 μM). Subsequent assay identified ethyl gallate as the best PPARγ antagonist (IC50 = 5.4 μM) among the hit compounds inhibiting PTPN6. Ethyl gallate upregulated glucose uptake and downregulated adipogenesis in 3T3-L1 cells as anticipated. These results strongly suggest that ethyl gallate, which targets both PTPN6 and PPARγ, is a potent therapeutic candidate to combat diabesity.
Collapse
Affiliation(s)
- Dohee Ahn
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea; (D.A.); (J.K.); (G.N.); (X.Z.); (J.Y.H.); (J.K.K.)
| | - Jinsoo Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea; (D.A.); (J.K.); (G.N.); (X.Z.); (J.Y.H.); (J.K.K.)
| | - Gibeom Nam
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea; (D.A.); (J.K.); (G.N.); (X.Z.); (J.Y.H.); (J.K.K.)
| | - Xiaodi Zhao
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea; (D.A.); (J.K.); (G.N.); (X.Z.); (J.Y.H.); (J.K.K.)
| | - Jihee Kwon
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Korea;
| | - Ji Young Hwang
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea; (D.A.); (J.K.); (G.N.); (X.Z.); (J.Y.H.); (J.K.K.)
| | - Jae Kwan Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea; (D.A.); (J.K.); (G.N.); (X.Z.); (J.Y.H.); (J.K.K.)
| | - Sun-Young Yoon
- Department of Cosmetic Science, Kwangju Women’s University, Gwangju 62396, Korea;
| | - Sang J. Chung
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea; (D.A.); (J.K.); (G.N.); (X.Z.); (J.Y.H.); (J.K.K.)
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Korea;
- Correspondence: ; Tel.: +82-31-290-7703
| |
Collapse
|
24
|
Kirschner KM, Foryst-Ludwig A, Gohlke S, Li C, Flores RE, Kintscher U, Schupp M, Schulz TJ, Scholz H. Wt1 haploinsufficiency induces browning of epididymal fat and alleviates metabolic dysfunction in mice on high-fat diet. Diabetologia 2022; 65:528-540. [PMID: 34846543 PMCID: PMC8803700 DOI: 10.1007/s00125-021-05621-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 09/24/2021] [Indexed: 11/26/2022]
Abstract
AIMS/HYPOTHESIS Despite a similar fat storing function, visceral (intra-abdominal) white adipose tissue (WAT) is detrimental, whereas subcutaneous WAT is considered to protect against metabolic disease. Recent findings indicate that thermogenic genes, expressed in brown adipose tissue (BAT), can be induced primarily in subcutaneous WAT. Here, we investigate the hypothesis that the Wilms tumour gene product (WT1), which is expressed in intra-abdominal WAT but not in subcutaneous WAT and BAT, suppresses a thermogenic program in white fat cells. METHODS Heterozygous Wt1 knockout mice and their wild-type littermates were examined in terms of thermogenic and adipocyte-selective gene expression. Glucose tolerance and hepatic lipid accumulation in these mice were assessed under normal chow and high-fat diet conditions. Pre-adipocytes isolated from the stromal vascular fraction of BAT were transduced with Wt1-expressing retrovirus, induced to differentiate and analysed for the expression of thermogenic and adipocyte-selective genes. RESULTS Expression of the thermogenic genes Cpt1b and Tmem26 was enhanced and transcript levels of Ucp1 were on average more than tenfold higher in epididymal WAT of heterozygous Wt1 knockout mice compared with wild-type mice. Wt1 heterozygosity reduced epididymal WAT mass, improved whole-body glucose tolerance and alleviated severe hepatic steatosis upon diet-induced obesity in mice. Retroviral expression of WT1 in brown pre-adipocytes, which lack endogenous WT1, reduced mRNA levels of Ucp1, Ppargc1a, Cidea, Prdm16 and Cpt1b upon in vitro differentiation by 60-90%. WT1 knockdown in epididymal pre-adipocytes significantly lowered Aldh1a1 and Zfp423 transcripts, two key suppressors of the thermogenic program. Conversely, Aldh1a1 and Zfp423 mRNA levels were increased approximately five- and threefold, respectively, by retroviral expression of WT1 in brown pre-adipocytes. CONCLUSION/INTERPRETATION WT1 functions as a white adipocyte determination factor in epididymal WAT by suppressing thermogenic genes. Reducing Wt1 expression in this and other intra-abdominal fat depots may represent a novel treatment strategy in metabolic disease.
Collapse
Affiliation(s)
- Karin M Kirschner
- Institut für Vegetative Physiologie, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Anna Foryst-Ludwig
- Institut für Pharmakologie, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Sabrina Gohlke
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
| | - Chen Li
- Institut für Pharmakologie, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Roberto E Flores
- Institut für Pharmakologie, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ulrich Kintscher
- Institut für Pharmakologie, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Michael Schupp
- Institut für Pharmakologie, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Tim J Schulz
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, Potsdam-Rehbrücke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Holger Scholz
- Institut für Vegetative Physiologie, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
| |
Collapse
|
25
|
Althubiti M. Tyrosine kinase targeting: A potential therapeutic strategy for diabetes. SAUDI JOURNAL OF MEDICINE AND MEDICAL SCIENCES 2022; 10:183-191. [PMID: 36247049 PMCID: PMC9555044 DOI: 10.4103/sjmms.sjmms_492_21] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 12/06/2021] [Accepted: 08/11/2022] [Indexed: 12/01/2022] Open
Abstract
Tyrosine kinase inhibitors (TKIs) have been studied extensively in cancer research, ultimately resulting in the approval of many drugs for cancer therapy. Recent evidence from reported clinical cases and experimental studies have suggested that some of these drugs have a potential role in diabetes treatment. These TKIs include imatinib, sunitinib, dasatinib, erlotinib, nilotinib, neratinib, and ibrutinib. As a result of promising findings, imatinib has been used in a phase II clinical trial. In this review, studies that used TKIs in the treatment of both types of diabetes are critically discussed. In addition, the different molecular mechanisms of action of these drugs in diabetes models are also highlighted to understand their antidiabetic mode of action.
Collapse
|
26
|
Welsh N. Are off-target effects of imatinib the key to improving beta-cell function in diabetes? Ups J Med Sci 2022; 127:8841. [PMID: 36187072 PMCID: PMC9487420 DOI: 10.48101/ujms.v127.8841] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/18/2022] [Accepted: 07/27/2022] [Indexed: 11/18/2022] Open
Abstract
The small tyrosine kinase (TK) inhibitor imatinib mesylate (Gleevec, STI571) protects against both type 1 and type 2 diabetes, but as it inhibits many TKs and other proteins, it is not clear by which mechanisms it acts. This present review will focus on the possibility that imatinib acts, at least in part, by improving beta-cell function and survival via off-target effects on beta-cell signaling/metabolic flow events. Particular attention will be given to the possibility that imatinib and other TK inhibitors function as inhibitors of mitochondrial respiration. A better understanding of how imatinib counteracts diabetes will possibly help to clarify the pathogenic role of beta-cell signaling events and mitochondrial function, and hopefully leading to improved treatment of the disease.
Collapse
Affiliation(s)
- Nils Welsh
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
27
|
Toobian D, Ghosh P, Katkar GD. Parsing the Role of PPARs in Macrophage Processes. Front Immunol 2021; 12:783780. [PMID: 35003101 PMCID: PMC8727354 DOI: 10.3389/fimmu.2021.783780] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022] Open
Abstract
Cells are richly equipped with nuclear receptors, which act as ligand-regulated transcription factors. Peroxisome proliferator activated receptors (PPARs), members of the nuclear receptor family, have been extensively studied for their roles in development, differentiation, and homeostatic processes. In the recent past, there has been substantial interest in understanding and defining the functions of PPARs and their agonists in regulating innate and adaptive immune responses as well as their pharmacologic potential in combating acute and chronic inflammatory disease. In this review, we focus on emerging evidence of the potential roles of the PPAR subtypes in macrophage biology. We also discuss the roles of dual and pan PPAR agonists as modulators of immune cell function, microbial infection, and inflammatory diseases.
Collapse
Affiliation(s)
- Daniel Toobian
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, CA, United States
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, CA, United States
- Rebecca and John Moore Comprehensive Cancer Center, University of California San Diego, San Diego, CA, United States
- Department of Medicine, University of California San Diego, San Diego, CA, United States
- Veterans Affairs Medical Center, La Jolla, CA, United States
| | - Gajanan D. Katkar
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, CA, United States
| |
Collapse
|
28
|
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: 2.3] [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
|
29
|
Sivamani Y, Shanmugarajan D, Durai Ananda Kumar T, Faizan S, Channappa B, Naishima NL, Prashantha Kumar BR. A promising in silico protocol to develop novel PPARγ antagonists as potential anticancer agents: Design, synthesis and experimental validation via PPARγ protein activity and competitive binding assay. Comput Biol Chem 2021; 95:107600. [PMID: 34794076 DOI: 10.1016/j.compbiolchem.2021.107600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 10/29/2021] [Accepted: 11/04/2021] [Indexed: 01/11/2023]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ), a member of the nuclear receptor superfamily is an excellent example of targets that orchestrates cancer, inflammation, lipid and glucose metabolism. We report a protocol for the development of novel PPARγ antagonists by employing 3D QSAR based virtual screening for the identification of ligands with anticancer properties. The models are generated based on a large and diverse set of PPARγ antagonist ligands by the HYPOGEN algorithm using Discovery Studio 2019 drug design software. Among the 10 hypotheses generated, Hypotheses 2 showed the highest correlation coefficient values of 0.95 with less RMS deviation of 1.193. Validation of the developed pharmacophore model was performed by Fischer's randomization and screening against test and decoy set. The GH score or goodness score was found to be 0.81 indicating moderate to a good model. The selected pharmacophore model Hypo 2 was used as a query model for further screening of 11,145 compounds from the PubChem, sc-PDB structure database, and designed novel ligands. Based on fit values and ADMET filter, the final 10 compounds with the predicated activity of ≤ 3 nM were subjected for docking analysis. Docking analysis revealed the unique binding mode with hydrophobic amino acid that can cause destabilization of the H12 which is an important molecular mechanism to prove its antagonist action. Based on high CDocker scores, Cpd31 was synthesized, purified, analyzed and screened for PPARγ competitive binding by TR-FRET assay. The biochemical protein binding results matched the predicted results. Further, Cpd31 was screened against cancer cells and validated the results.
Collapse
Affiliation(s)
- Yuvaraj Sivamani
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570 015, Karnataka, India
| | - Dhivya Shanmugarajan
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570 015, Karnataka, India
| | - T Durai Ananda Kumar
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570 015, Karnataka, India
| | - Syed Faizan
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570 015, Karnataka, India
| | - Bhavya Channappa
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570 015, Karnataka, India
| | - Namburu Lalitha Naishima
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570 015, Karnataka, India
| | - B R Prashantha Kumar
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570 015, Karnataka, India.
| |
Collapse
|
30
|
Human Brown Adipose Tissue and Metabolic Health: Potential for Therapeutic Avenues. Cells 2021; 10:cells10113030. [PMID: 34831253 PMCID: PMC8616549 DOI: 10.3390/cells10113030] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 12/31/2022] Open
Abstract
Obesity-associated metabolic abnormalities comprise a cluster of conditions including dyslipidemia, insulin resistance, diabetes and cardiovascular diseases that has affected more than 650 million people all over the globe. Obesity results from the accumulation of white adipose tissues mainly due to the chronic imbalance of energy intake and energy expenditure. A variety of approaches to treat or prevent obesity, including lifestyle interventions, surgical weight loss procedures and pharmacological approaches to reduce energy intake and increase energy expenditure have failed to substantially decrease the prevalence of obesity. Brown adipose tissue (BAT), the primary source of thermogenesis in infants and small mammals may represent a promising therapeutic target to treat obesity by promoting energy expenditure through non-shivering thermogenesis mediated by mitochondrial uncoupling protein 1 (UCP1). Since the confirmation of functional BAT in adult humans by several groups, approximately a decade ago, and its association with a favorable metabolic phenotype, intense interest on the significance of BAT in adult human physiology and metabolic health has emerged within the scientific community to explore its therapeutic potential for the treatment of obesity and metabolic diseases. A substantially decreased BAT activity in individuals with obesity indicates a role for BAT in the setting of human obesity. On the other hand, BAT mass and its prevalence correlate with lower body mass index (BMI), decreased age and lower glucose levels, leading to a lower incidence of cardio-metabolic diseases. The increased cold exposure in adult humans with undetectable BAT was associated with decreased body fat mass and increased insulin sensitivity. A deeper understanding of the role of BAT in human metabolic health and its interrelationship with body fat distribution and deciphering proper strategies to increase energy expenditure, by either increasing functional BAT mass or inducing white adipose browning, holds the promise for possible therapeutic avenues for the treatment of obesity and associated metabolic disorders.
Collapse
|
31
|
Herlea-Pana O, Eeda V, Undi RB, Lim HY, Wang W. Pharmacological Inhibition of Inositol-Requiring Enzyme 1α RNase Activity Protects Pancreatic Beta Cell and Improves Diabetic Condition in Insulin Mutation-Induced Diabetes. Front Endocrinol (Lausanne) 2021; 12:749879. [PMID: 34675883 PMCID: PMC8524045 DOI: 10.3389/fendo.2021.749879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/20/2021] [Indexed: 12/25/2022] Open
Abstract
β-cell ER stress plays an important role in β-cell dysfunction and death during the pathogenesis of diabetes. Proinsulin misfolding is regarded as one of the primary initiating factors of ER stress and unfolded protein response (UPR) activation in β-cells. Here, we found that the ER stress sensor inositol-requiring enzyme 1α (IRE1α) was activated in the Akita mice, a mouse model of mutant insulin gene-induced diabetes of youth (MIDY), a monogenic diabetes. Normalization of IRE1α RNase hyperactivity by pharmacological inhibitors significantly ameliorated the hyperglycemic conditions and increased serum insulin levels in Akita mice. These benefits were accompanied by a concomitant protection of functional β-cell mass, as shown by the suppression of β-cell apoptosis, increase in mature insulin production and reduction of proinsulin level. At the molecular level, we observed that the expression of genes associated with β-cell identity and function was significantly up-regulated and ER stress and its associated inflammation and oxidative stress were suppressed in islets from Akita mice treated with IRE1α RNase inhibitors. This study provides the evidence of the in vivo efficacy of IRE1α RNase inhibitors in Akita mice, pointing to the possibility of targeting IRE1α RNase as a therapeutic direction for the treatment of diabetes.
Collapse
Affiliation(s)
- Oana Herlea-Pana
- Department of Medicine, Division of Endocrinology, Harold Hamm Diabetes Center, Oklahoma City, OK, United States
| | - Venkateswararao Eeda
- Department of Medicine, Division of Endocrinology, Harold Hamm Diabetes Center, Oklahoma City, OK, United States
| | - Ram Babu Undi
- Department of Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science Center, Oklahoma City, OK, United States
| | - Hui-Ying Lim
- Department of Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science Center, Oklahoma City, OK, United States
| | - Weidong Wang
- Department of Medicine, Division of Endocrinology, Harold Hamm Diabetes Center, Oklahoma City, OK, United States
| |
Collapse
|
32
|
Gitelman SE, Bundy BN, Ferrannini E, Lim N, Blanchfield JL, DiMeglio LA, Felner EI, Gaglia JL, Gottlieb PA, Long SA, Mari A, Mirmira RG, Raskin P, Sanda S, Tsalikian E, Wentworth JM, Willi SM, Krischer JP, Bluestone JA. Imatinib therapy for patients with recent-onset type 1 diabetes: a multicentre, randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Diabetes Endocrinol 2021; 9:502-514. [PMID: 34214479 PMCID: PMC8494464 DOI: 10.1016/s2213-8587(21)00139-x] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/06/2021] [Accepted: 05/10/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Type 1 diabetes results from autoimmune-mediated destruction of β cells. The tyrosine kinase inhibitor imatinib might affect relevant immunological and metabolic pathways, and preclinical studies show that it reverses and prevents diabetes. Our aim was to evaluate the safety and efficacy of imatinib in preserving β-cell function in patients with recent-onset type 1 diabetes. METHODS We did a multicentre, randomised, double-blind, placebo-controlled, phase 2 trial. Patients with recent-onset type 1 diabetes (<100 days from diagnosis), aged 18-45 years, positive for at least one type of diabetes-associated autoantibody, and with a peak stimulated C-peptide of greater than 0·2 nmol L-1 on a mixed meal tolerance test (MMTT) were enrolled from nine medical centres in the USA (n=8) and Australia (n=1). Participants were randomly assigned (2:1) to receive either 400 mg imatinib mesylate (4 × 100 mg film-coated tablets per day) or matching placebo for 26 weeks via a computer-generated blocked randomisation scheme stratified by centre. Treatment assignments were masked for all participants and study personnel except pharmacists at each clinical site. The primary endpoint was the difference in the area under the curve (AUC) mean for C-peptide response in the first 2 h of an MMTT at 12 months in the imatinib group versus the placebo group, with use of an ANCOVA model adjusting for sex, baseline age, and baseline C-peptide, with further observation up to 24 months. The primary analysis was by intention to treat (ITT). Safety was assessed in all randomly assigned participants. This study is registered with ClinicalTrials.gov, NCT01781975 (completed). FINDINGS Patients were screened and enrolled between Feb 12, 2014, and May 19, 2016. 45 patients were assigned to receive imatinib and 22 to receive placebo. After withdrawals, 43 participants in the imatinib group and 21 in the placebo group were included in the primary ITT analysis at 12 months. The study met its primary endpoint: the adjusted mean difference in 2-h C-peptide AUC at 12 months for imatinib versus placebo treatment was 0·095 (90% CI -0·003 to 0·191; p=0·048, one-tailed test). This effect was not sustained out to 24 months. During the 24-month follow-up, 32 (71%) of 45 participants who received imatinib had a grade 2 severity or worse adverse event, compared with 13 (59%) of 22 participants who received placebo. The most common adverse events (grade 2 severity or worse) that differed between the groups were gastrointestinal issues (six [13%] participants in the imatinib group, primarily nausea, and none in the placebo group) and additional laboratory investigations (ten [22%] participants in the imatinib group and two [9%] in the placebo group). Per the trial protocol, 17 (38%) participants in the imatinib group required a temporary modification in drug dosing and six (13%) permanently discontinued imatinib due to adverse events; five (23%) participants in the placebo group had temporary modifications in dosing and none had a permanent discontinuation due to adverse events. INTERPRETATION A 26-week course of imatinib preserved β-cell function at 12 months in adults with recent-onset type 1 diabetes. Imatinib might offer a novel means to alter the course of type 1 diabetes. Future considerations are defining ideal dose and duration of therapy, safety and efficacy in children, combination use with a complimentary drug, and ability of imatinib to delay or prevent progression to diabetes in an at-risk population; however, careful monitoring for possible toxicities is required. FUNDING Juvenile Research Diabetes Foundation.
Collapse
Affiliation(s)
| | | | | | - Noha Lim
- Immune Tolerance Network, Bethesda, MD, USA
| | | | | | | | - Jason L Gaglia
- Section on Immunology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | | | | | - Andrea Mari
- CNR Institute of Neurosciences, Padua, Italy
| | | | - Philip Raskin
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Srinath Sanda
- University of California San Francisco, San Francisco, CA, USA
| | | | - John M Wentworth
- Walter and Eliza Hall Institute and Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Steven M Willi
- Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | |
Collapse
|
33
|
Frkic RL, Richter K, Bruning JB. The therapeutic potential of inhibiting PPARγ phosphorylation to treat type 2 diabetes. J Biol Chem 2021; 297:101030. [PMID: 34339734 PMCID: PMC8387755 DOI: 10.1016/j.jbc.2021.101030] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 11/30/2022] Open
Abstract
A promising approach for treating type 2 diabetes mellitus (T2DM) is to target the Peroxisome Proliferator-Activated Receptor γ (PPARγ) transcription factor, which regulates the expression of proteins critical for T2DM. Mechanisms involved in PPARγ signaling are poorly understood, yet globally increasing T2DM prevalence demands improvements in drug design. Synthetic, nonactivating PPARγ ligands can abolish the phosphorylation of PPARγ at Ser273, a posttranslational modification correlated with obesity and insulin resistance. It is not understood how these ligands prevent phosphorylation, and the lack of experimental mechanistic information can be attributed to previous ambiguity in the field as well as to limitations in experimental approaches; in silico modeling currently provides the only insight into how ligands block Ser273 phosphorylation. The future availability of experimental evidence is critical for clarifying the mechanism by which ligands prevent phosphorylation and should be the priority of future T2DM-focused research. Following this, the properties of ligands that enable them to block phosphorylation can be improved upon to generate ligands tailored for blocking phosphorylation and therefore restoring insulin sensitivity. This would represent a significant step forward for treating T2DM. This review summarizes current knowledge of the roles of PPARγ in T2DM as well as the effects of synthetic ligands on the modulation of these roles. We hypothesize potential factors that contribute to the reduction in recent developments and summarize what has currently been done to shed light on this critical field of research.
Collapse
Affiliation(s)
- Rebecca L Frkic
- The Institute for Photonics and Advanced Sensing, and School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Katharina Richter
- Richter Lab, Department of Surgery, Basil Hetzel Institute for Translational Health Research, The University of Adelaide, Adelaide, South Australia, Australia
| | - John B Bruning
- The Institute for Photonics and Advanced Sensing, and School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia.
| |
Collapse
|
34
|
Sert G, Kucukguven A, Zırh EB, Demirtaş TT, Çakar AN, Gümüşderelioğlu M, Calis M. Photobiomodulation with polychromatic light (600-1200 nm) improves fat graft survival by increasing adipocyte viability, neovascularization, and reducing inflammation in a rat model. Lasers Surg Med 2021; 54:268-280. [PMID: 34289510 DOI: 10.1002/lsm.23457] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/03/2021] [Indexed: 01/11/2023]
Abstract
OBJECTIVES Unpredictability with the final volume and viability of the graft are the major concerns in fat grafting. An experimental study was conducted to increase graft retention using photobiomodulation (PBM) with polychromatic light in near-infrared region (600-1200 nm) by utilizing its stimulatory effects on angiogenesis, neovascularization, adipocyte viability, and anti-inflammatory properties. METHODS A total of 24 rats were divided into four groups (n = 6) according to the applied polychromatic light protocol to the recipient site (none, before fat transfer, after fat transfer, and combined). In all groups, inguinal fat pad was excised, measured for volume and weight, and transferred to the dorsum of the rat. At the end of the experiment, fat grafts were harvested from the recipient site for volume and weight measurements, histological, and immunohistochemical evaluation. RESULTS Intergroup comparison revealed that fat graft retention regarding weight and volume, was significantly superior in Group IV (p = 0.049 and p = 0.043, respectively), which polychromatic light was applied both before and after transfer of the graft. Hematoxylin-eosin and Masson's trichrome stained sections showed absence of necrosis, fibrosis, inflammation, cyst formation, and increased vascularization of both inner and outer zones of the grafts in Group IV. Also, immunohistochemical staining scores for perilipin (indicator for adipocyte viability), CD31 and VEGF (indicators for angiogenesis and neovascularization) were significantly higher (p < 0.001). Ki67 scores were significantly lower in this group because of anti-inflammatory environment (p < 0.001). CONCLUSIONS Application of PBM to the recipient site before and after fat transfer improved outcomes in rats at 56 day after fat grafting by means of volume retention, increased neovascularization and adipocyte viability and reduced necrosis, fibrosis and inflammation.
Collapse
Affiliation(s)
- Gokhan Sert
- Department of Plastic Reconstructive and Aesthetic Surgery, Faculty of Medicine, Hacettepe University Ankara, Ankara, Turkey
| | - Arda Kucukguven
- Department of Plastic Reconstructive and Aesthetic Surgery, Faculty of Medicine, Hacettepe University Ankara, Ankara, Turkey
| | - Elham Bahador Zırh
- Department of Histology and Embryology, Faculty of Medicine, TOBB ETU University, Ankara, Turkey
| | - Tugrul T Demirtaş
- Department of Bioengineering, Faculty of Engineering, Hacettepe University, Ankara, Turkey.,Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Erciyes University, Kayseri, Turkey
| | - Ayşe Nur Çakar
- Department of Histology and Embryology, Faculty of Medicine, TOBB ETU University, Ankara, Turkey
| | - Menemşe Gümüşderelioğlu
- Department of Bioengineering, Faculty of Engineering, Hacettepe University, Ankara, Turkey.,Department of Chemical Engineering, Faculty of Engineering, Hacettepe University, Ankara, Turkey
| | - Mert Calis
- Department of Plastic Reconstructive and Aesthetic Surgery, Faculty of Medicine, Hacettepe University Ankara, Ankara, Turkey
| |
Collapse
|
35
|
Zhang Y, Bobe G, Miranda CL, Lowry MB, Hsu VL, Lohr CV, Wong CP, Jump DB, Robinson MM, Sharpton TJ, Maier CS, Stevens JF, Gombart AF. Tetrahydroxanthohumol, a xanthohumol derivative, attenuates high-fat diet-induced hepatic steatosis by antagonizing PPARγ. eLife 2021; 10:e66398. [PMID: 34128467 PMCID: PMC8205491 DOI: 10.7554/elife.66398] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 05/18/2021] [Indexed: 12/13/2022] Open
Abstract
We previously reported xanthohumol (XN), and its synthetic derivative tetrahydro-XN (TXN), attenuates high-fat diet (HFD)-induced obesity and metabolic syndrome in C57Bl/6J mice. The objective of the current study was to determine the effect of XN and TXN on lipid accumulation in the liver. Non-supplemented mice were unable to adapt their caloric intake to 60% HFD, resulting in obesity and hepatic steatosis; however, TXN reduced weight gain and decreased hepatic steatosis. Liver transcriptomics indicated that TXN might antagonize lipogenic PPARγ actions in vivo. XN and TXN inhibited rosiglitazone-induced 3T3-L1 cell differentiation concomitant with decreased expression of lipogenesis-related genes. A peroxisome proliferator activated receptor gamma (PPARγ) competitive binding assay showed that XN and TXN bind to PPARγ with an IC50 similar to pioglitazone and 8-10 times stronger than oleate. Molecular docking simulations demonstrated that XN and TXN bind in the PPARγ ligand-binding domain pocket. Our findings are consistent with XN and TXN acting as antagonists of PPARγ.
Collapse
Affiliation(s)
- Yang Zhang
- School of Biological and Population Health Sciences, Nutrition Program, Linus Pauling Institute, Oregon State UniversityCorvallisUnited States
| | - Gerd Bobe
- Department of Animal Sciences, Linus Pauling Institute, Oregon State UniversityCorvallisUnited States
| | - Cristobal L Miranda
- Department of Pharmaceutical Sciences, Linus Pauling Institute, Oregon State UniversityCorvallisUnited States
| | - Malcolm B Lowry
- Department of Microbiology, Oregon State UniversityCorvallisUnited States
| | - Victor L Hsu
- Department of Biochemistry and Biophysics, Oregon State UniversityCorvallisUnited States
| | - Christiane V Lohr
- Department of Biomedical Science, Carlson College of Veterinary MedicineCorvallisUnited States
| | - Carmen P Wong
- School of Biological and Population Health Sciences, Nutrition Program, Linus Pauling Institute, Oregon State UniversityCorvallisUnited States
| | - Donald B Jump
- School of Biological and Population Health Sciences, Nutrition Program, Linus Pauling Institute, Oregon State UniversityCorvallisUnited States
| | - Matthew M Robinson
- School of Biological and Population Health Sciences, Kinesiology Program, Oregon State UniversityCorvallisUnited States
| | - Thomas J Sharpton
- Department of Microbiology, Department of Statistics, Oregon State UniversityCorvallisUnited States
| | - Claudia S Maier
- Department of Chemistry, Linus Pauling Institute, Oregon State UniversityCorvallisUnited States
| | - Jan F Stevens
- Department of Pharmaceutical Sciences, Linus Pauling Institute, Oregon State UniversityCorvallisUnited States
| | - Adrian F Gombart
- Linus Pauling Institute, Department of Biochemistry and Biophysics, Oregon State UniversityCorvallisUnited States
| |
Collapse
|
36
|
Dai J, Huang X, Zhang C, Luo X, Cao S, Wang J, Liu B, Gao J. Berberine regulates lipid metabolism via miR-192 in porcine oocytes matured in vitro. Vet Med Sci 2021; 7:950-959. [PMID: 33818910 PMCID: PMC8136937 DOI: 10.1002/vms3.393] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 10/07/2020] [Accepted: 10/25/2020] [Indexed: 12/26/2022] Open
Abstract
Background The berberine (Ber) is an isoquinoline alkaloid compound extracted from Rhizoma coptidis and has the effect that reduces adipose. MicroRNA‐192 (miR‐192) is related to fat metabolism. However, the relevant mechanism of berberine on lipid metabolism during in vitro maturation (IVM) of porcine oocytes remains unclear. Objectives In this study, we investigated the molecular mechanism by which berberine promotes the IVM and lipid metabolism of porcine oocytes via miR‐192. Methods Ber was added to IVM medium of porcine oocytes. MiR‐192 agomir, miR‐192 antagomir and negative control fragment were microinjected into the cytoplasm of oocytes without Ber. Rates of oocyte IVM and embryonic development in each group were observed. The content of lipid droplets in IVM oocytes in each group was analyzed by Nile red staining. Expression levels of miR‐192 and FABP3, SREBF1 and PPARG, were detected by qPCR and western blotting. The target genes of miR‐192 were determined by luciferase reporter assays. Results and Conclusions We found that Ber significantly increased the rate of oocytes IVM and blastocyst development, and decreased the area and numbers of lipid droplets in IVM oocytes. Ber significantly increased the expression of miR‐192 in IVM oocytes, and significantly decreased the expression of SREBF1 and PPARG, which were target genes of miR‐192. This study indicates that Ber promotes lipid metabolism in porcine oocytes by activating the expression of miR‐192 and down‐regulating SREBF1 and PPARG, thus, improving IVM of porcine oocytes.
Collapse
Affiliation(s)
- JiaGe Dai
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - XiaoMeng Huang
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Chao Zhang
- College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - XiaoFei Luo
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - SuYing Cao
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - JunLi Wang
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Bing Liu
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - JianMing Gao
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| |
Collapse
|
37
|
Verma R, Choi D, Chen AJ, Harrington CA, Wilson DJ, Grossniklaus HE, Dailey RA, Ng J, Steele EA, Planck SR, Korn BS, Kikkawa D, Czyz CN, Foster JA, Kazim M, Harris GJ, Edward DP, Al-Hussain H, Maktabi AMY, Alabiad C, Garcia A, Rosenbaum JT. Enrichment of IGF-1R and PPARγ signalling pathways in orbital inflammatory diseases: steps toward understanding pathogenesis. Br J Ophthalmol 2021; 106:1012-1017. [PMID: 33637620 DOI: 10.1136/bjophthalmol-2020-318330] [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: 11/02/2020] [Revised: 01/21/2021] [Accepted: 02/05/2021] [Indexed: 11/03/2022]
Abstract
BACKGROUND Orbital inflammatory disease (OID) encompasses a wide range of pathology including thyroid-associated orbitopathy (TAO), granulomatosis with polyangiitis (GPA), sarcoidosis and non-specific orbital inflammation (NSOI), accounting for up to 6% of orbital diseases. Understanding the underlying pathophysiology of OID can improve diagnosis and help target therapy. AIMS To test the hypothesis that shared signalling pathways are activated in different forms of OID. METHODS In this secondary analysis, pathway analysis was performed on the previously reported differentially expressed genes from orbital adipose tissue using patients with OID and healthy controls who were characterised by microarray. For the original publications, tissue specimens were collected from oculoplastic surgeons at 10 international centres representing four countries (USA, Canada, Australia and Saudi Arabia). Diagnoses were independently confirmed by two masked ocular pathologists (DJW, HEG). Gene expression profiling analysis was performed at the Oregon Health & Science University. Eighty-three participants were included: 25 with TAO, 6 with orbital GPA, 7 with orbital sarcoidosis, 25 with NSOI and 20 healthy controls. RESULTS Among the 83 subjects (mean (SD) age, 52.8 (18.3) years; 70% (n=58) female), those with OID demonstrated perturbation of the downstream gene expressions of the IGF-1R (MAPK/RAS/RAF/MEK/ERK and PI3K/Akt/mTOR pathways), peroxisome proliferator-activated receptor-γ (PPARγ), adipocytokine and AMPK signalling pathways compared with healthy controls. Specifically, GPA samples differed from controls in gene expression within the insulin-like growth factor-1 receptor (IGF-1R, PI3K-Akt (p=0.001), RAS (p=0.005)), PPARγ (p=0.002), adipocytokine (p=0.004) or AMPK (p=<0.001) pathways. TAO, sarcoidosis and NSOI samples were also found to have statistically significant differential gene expression in these pathways. CONCLUSIONS Although OID includes a heterogenous group of pathologies, TAO, GPA, sarcoidosis and NSOI share enrichment of common gene signalling pathways, namely IGF-1R, PPARγ, adipocytokine and AMPK. Pathway analyses of gene expression suggest that other forms of orbital inflammation in addition to TAO may benefit from blockade of IGF-1R signalling pathways.
Collapse
Affiliation(s)
- Rohan Verma
- Oculofacial Plastic and Reconstructive Surgery, Oregon Health & Science University Casey Eye Institute, Portland, Oregon, USA.,Casey Eye Institute, Oregon Health & Science University Casey Eye Institute, Portland, Oregon, USA
| | - Dongseok Choi
- Casey Eye Institute, Oregon Health & Science University Casey Eye Institute, Portland, Oregon, USA.,OHSU-PSU School of Public Health, Oregon Health & Science University, Portland, Oregon, USA.,Graduate School of Dentistry, Kyung Hee University, Seoul, South Korea.,Department of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Allison J Chen
- Oculofacial Plastic and Reconstructive Surgery, University of California San Diego- Shiley Eye Institute and Viterbi Family Department of Ophthalmology, La Jolla, California, USA
| | - Christina A Harrington
- Integrated Genomics Laboratory, Oregon Health & Science University, Portland, Oregon, USA
| | - David J Wilson
- Casey Eye Institute, Oregon Health & Science University Casey Eye Institute, Portland, Oregon, USA
| | | | - Roger A Dailey
- Oculofacial Plastic and Reconstructive Surgery, Oregon Health & Science University Casey Eye Institute, Portland, Oregon, USA.,Casey Eye Institute, Oregon Health & Science University Casey Eye Institute, Portland, Oregon, USA
| | - John Ng
- Oculofacial Plastic and Reconstructive Surgery, Oregon Health & Science University Casey Eye Institute, Portland, Oregon, USA.,Casey Eye Institute, Oregon Health & Science University Casey Eye Institute, Portland, Oregon, USA
| | - Eric A Steele
- Oculofacial Plastic and Reconstructive Surgery, Oregon Health & Science University Casey Eye Institute, Portland, Oregon, USA.,Casey Eye Institute, Oregon Health & Science University Casey Eye Institute, Portland, Oregon, USA
| | - Stephen R Planck
- Casey Eye Institute, Oregon Health & Science University Casey Eye Institute, Portland, Oregon, USA.,Devers Eye Institute, Legacy Health System, Portland, Oregon, USA
| | - Bobby S Korn
- Oculofacial Plastic and Reconstructive Surgery, University of California San Diego- Shiley Eye Institute and Viterbi Family Department of Ophthalmology, La Jolla, California, USA
| | - Don Kikkawa
- Oculofacial Plastic and Reconstructive Surgery, University of California San Diego- Shiley Eye Institute and Viterbi Family Department of Ophthalmology, La Jolla, California, USA
| | - Craig N Czyz
- Oculofacial Plastic and Reconstructive Surgery, Ohio Health, Columbus, Ohio, USA
| | - Jill A Foster
- Oculofacial Plastic and Reconstructive Surgery, The Ohio State University, Nationwide Children's Hospital, Ophthalmic Surgeons and Consultants of Ohio, Columbus, Ohio, USA
| | - Michael Kazim
- Edward S Harkness Eye Institute, Columbia University, New York, New York, USA
| | - Gerald J Harris
- Oculofacial Plastic and Reconstructive Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Deepak P Edward
- Ophthalmology and Visual Sciences, University of Illinois College of Medicine, Chicago, Illinois, USA.,Research Department, King Khaled Eye Specialist Hospital, Riyadh, Saudi Arabia
| | - Haila Al-Hussain
- Research Department, King Khaled Eye Specialist Hospital, Riyadh, Saudi Arabia
| | - Azza M Y Maktabi
- Research Department, King Khaled Eye Specialist Hospital, Riyadh, Saudi Arabia
| | - Chris Alabiad
- Oculofacial Plastic and Reconstructive Surgery, University of Miami Health System Bascom Palmer Eye Institute, Miami, Florida, USA
| | - Armando Garcia
- Oculofacial Plastic and Reconstructive Surgery, University of Miami Health System Bascom Palmer Eye Institute, Miami, Florida, USA
| | - James T Rosenbaum
- Casey Eye Institute, Oregon Health & Science University Casey Eye Institute, Portland, Oregon, USA .,Department of Medicine, Oregon Health & Science University, Portland, Oregon, USA.,Devers Eye Institute, Legacy Health System, Portland, Oregon, USA
| |
Collapse
|
38
|
Shao M, Hepler C, Zhang Q, Shan B, Vishvanath L, Henry GH, Zhao S, An YA, Wu Y, Strand DW, Gupta RK. Pathologic HIF1α signaling drives adipose progenitor dysfunction in obesity. Cell Stem Cell 2021; 28:685-701.e7. [PMID: 33539723 DOI: 10.1016/j.stem.2020.12.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 10/19/2020] [Accepted: 12/15/2020] [Indexed: 01/06/2023]
Abstract
Adipose precursor cells (APCs) exhibit regional variation in response to obesity, for unclear reasons. Here, we reveal that HIFα-induced PDGFRβ signaling within murine white adipose tissue (WAT) PDGFRβ+ cells drives inhibitory serine 112 (S112) phosphorylation of PPARγ, the master regulator of adipogenesis. Levels of PPARγ S112 phosphorylation in WAT PDGFRβ+ cells are depot dependent, with levels of PPARγ phosphorylation in PDGFRβ+ cells inversely correlating with their capacity for adipogenesis upon high-fat-diet feeding. HIFα suppression in PDGFRβ+ progenitors promotes subcutaneous and intra-abdominal adipogenesis, healthy WAT remodeling, and improved metabolic health in obesity. These metabolic benefits are mimicked by treatment of obese mice with the PDGFR antagonist Imatinib, which promotes adipocyte hyperplasia and glucose tolerance in a progenitor cell PPARγ-dependent manner. Our studies unveil a mechanism underlying depot-specific responses of APCs to high-fat feeding and highlight the potential for APCs to be targeted pharmacologically to improve metabolic health in obesity.
Collapse
Affiliation(s)
- Mengle Shao
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chelsea Hepler
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Qianbin Zhang
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Bo Shan
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lavanya Vishvanath
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Gervaise H Henry
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shangang Zhao
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yu A An
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yibo Wu
- YCI Laboratory for Next-Generation Proteomics, RIKEN Center of Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Douglas W Strand
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rana K Gupta
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
39
|
Erucic Acid-Rich Yellow Mustard Oil Improves Insulin Resistance in KK-A y Mice. Molecules 2021; 26:molecules26030546. [PMID: 33494317 PMCID: PMC7864507 DOI: 10.3390/molecules26030546] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/07/2021] [Accepted: 01/16/2021] [Indexed: 12/24/2022] Open
Abstract
Obesity is a major risk factor for some metabolic disorders including type 2 diabetes. Enhancement of peroxisome proliferator-activated receptor (PPAR) γ, a master regulator of adipocyte differentiation, is known to increase insulin-sensitive small adipocytes. In contrast, decreased PPARγ activity is also reported to improve insulin resistance. We have previously identified erucic acid as a novel natural component suppressing PPARγ transcriptional activity. In this study, we investigated the effect of erucic acid-rich yellow mustard oil (YMO) on obese/diabetic KK-Ay mice. An in vitro luciferase reporter assay and mesenchymal stem cell (MSC) differentiation assay revealed that 25 µg/mL YMO significantly inhibited PPARγ transcriptional activity and differentiation of MSCs into adipocytes but promoted their differentiation into osteoblasts. In KK-Ay mice, dietary intake of 7.0% (w/w) YMO significantly decreased the surrogate indexes for insulin resistance and the infiltration of macrophages into adipose tissue. Furthermore, 7.0% YMO increased bone mineral density. These results suggest that YMO can ameliorate obesity-induced metabolic disorders.
Collapse
|
40
|
Das S, Chattopadhyay D, Chatterjee SK, Mondal SA, Majumdar SS, Mukhopadhyay S, Saha N, Velayutham R, Bhattacharya S, Mukherjee S. Increase in PPARγ inhibitory phosphorylation by Fetuin-A through the activation of Ras-MEK-ERK pathway causes insulin resistance. Biochim Biophys Acta Mol Basis Dis 2020; 1867:166050. [PMID: 33359696 DOI: 10.1016/j.bbadis.2020.166050] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 11/13/2020] [Accepted: 12/14/2020] [Indexed: 12/20/2022]
Abstract
Obesity induced insulin resistance is primarily regulated by the inhibitory phosphorylation of peroxisome proliferator-activated receptor γ at serine 273 (PPARγS273) which has been shown to be regulated by MEK and ERK. An upstream regulatory molecule of this pathway could be a therapeutic option. Here we analyzed the involvement of Fetuin-A (FetA), a key hepato-adipokine implicated in insulin resistance, as an upstream regulator molecule for the regulation of PPARγ inhibitory phosphorylation. Mice fed with standard diet (SD), high fat diet (HFD) and HFD with FetA knockdown (HFD-FetAKD) were used to examine the role of FetA on PPARγS273 phosphorylation in adipocytes. The mechanism of regulation and its effect on skeletal muscle were studied using primary adipocytes, 3T3-L1 (preadipocyte) and C2C12 (myotube) cell lines. Increased FetA in HFD mice strongly correlated with augmentation of PPARγS273 phosphorylation in inflamed adipocytes while knockdown of FetA suppressed it. This effect of FetA was mediated through the activation of Ras which in turn activated MEK and ERK. On addressing how FetA could stimulate activation of Ras, we found that FetA triggered TNFα in inflamed adipocytes which induced Ras activation. The ensuing sharp fall in adiponectin level attenuated AMPK activation in skeletal muscle cells affecting mitochondrial ATP production. Our data reveal the essential role of FetA induced activation of Ras in regulating PPARγ inhibitory phosphorylation through Ras-MEK-ERK pathway which downregulates adiponectin disrupting skeletal muscle mitochondrial bioenergetics. Thus, FetA mediated PPARγ inactivation has adverse consequences upon adipocyte-myocyte crosstalk leading to disruption of energy homeostasis and loss of insulin sensitivity.
Collapse
Affiliation(s)
- Snehasis Das
- Endocrinology and Metabolism Laboratory, Department of Zoology, Siksha Bhavana (Institute of Science), Visva-Bharati (A Central University), Santiniketan - 731235, India
| | - Dipanjan Chattopadhyay
- Endocrinology and Metabolism Laboratory, Department of Zoology, Siksha Bhavana (Institute of Science), Visva-Bharati (A Central University), Santiniketan - 731235, India
| | - Subhendu K Chatterjee
- Endocrinology and Metabolism Laboratory, Department of Zoology, Siksha Bhavana (Institute of Science), Visva-Bharati (A Central University), Santiniketan - 731235, India
| | - Samim Ali Mondal
- Department of Endocrinology & Metabolism, Institute of Post-Graduate Medical Education & Research-Seth Sukhlal Karnani Memorial (IPGME&R-SSKM) Hospital, Kolkata 700025, India
| | | | - Satinath Mukhopadhyay
- Department of Endocrinology & Metabolism, Institute of Post-Graduate Medical Education & Research-Seth Sukhlal Karnani Memorial (IPGME&R-SSKM) Hospital, Kolkata 700025, India
| | - Nirmalendu Saha
- Department of Zoology, North-Eastern Hill University, Shillong 793022, India
| | | | - Samir Bhattacharya
- Endocrinology and Metabolism Laboratory, Department of Zoology, Siksha Bhavana (Institute of Science), Visva-Bharati (A Central University), Santiniketan - 731235, India
| | - Sutapa Mukherjee
- Endocrinology and Metabolism Laboratory, Department of Zoology, Siksha Bhavana (Institute of Science), Visva-Bharati (A Central University), Santiniketan - 731235, India.
| |
Collapse
|
41
|
Pichavaram P, Shawky NM, Hartney TJ, Jun JY, Segar L. Imatinib improves insulin resistance and inhibits injury-induced neointimal hyperplasia in high fat diet-fed mice. Eur J Pharmacol 2020; 890:173666. [PMID: 33131722 DOI: 10.1016/j.ejphar.2020.173666] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/14/2020] [Accepted: 10/21/2020] [Indexed: 11/28/2022]
Abstract
Imatinib, a PDGF receptor tyrosine kinase inhibitor, has been shown to suppress intimal hyperplasia in different animal models under normal metabolic milieu, diabetic, and/or hypercholesterolemic conditions. However, the impact of imatinib treatment on injury-induced neointimal hyperplasia has not yet been investigated in the setting of insulin resistance without frank diabetes. Using a mouse model of high fat diet (HFD)-induced insulin resistance and guidewire-induced arterial injury, the present study demonstrates that intraperitoneal administration of imatinib (25 mg/kg/day) for ~3 weeks resulted in a marked attenuation of neointimal hyperplasia (intima/media ratio) by ~78% (n = 6-9 per group; P < 0.05). Imatinib treatment also led to significant improvements in key metabolic parameters. In particular, imatinib improved insulin resistance and glucose tolerance, as revealed by complete inhibition of HFD-induced increase in HOMA-IR index and AUCIPGTT, respectively. In addition, imatinib treatment led to diminutions in HFD-induced increases in plasma total cholesterol and triglycerides by ~73% and ~59%, respectively. Furthermore, imatinib decreased HFD-induced increase in visceral fat accumulation by ~51% (as determined by epididymal white adipose tissue weight). Importantly, imatinib treatment in HFD-fed mice enhanced plasma levels of high-molecular-weight adiponectin by ~2-fold without affecting total adiponectin. However, there were no significant changes in mean arterial pressure in insulin-resistant state or after imatinib exposure, as measured by tail-cuff method. Together, the present findings suggest that targeting PDGF receptor tyrosine kinase using imatinib may provide a realistic treatment option to prevent injury-induced neointimal hyperplasia and diet-induced insulin resistance in obesity.
Collapse
Affiliation(s)
- Prahalathan Pichavaram
- Charlie Norwood VA Medical Center, Augusta, GA, USA; Center for Pharmacy and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, USA
| | - Noha M Shawky
- Charlie Norwood VA Medical Center, Augusta, GA, USA; Center for Pharmacy and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, USA; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Egypt
| | | | - John Y Jun
- Division of Endocrinology, Diabetes, and Metabolism, Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Lakshman Segar
- Charlie Norwood VA Medical Center, Augusta, GA, USA; Center for Pharmacy and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, USA; Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA; Vascular Biology Center, Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA.
| |
Collapse
|
42
|
Kuryłowicz A, Puzianowska-Kuźnicka M. Induction of Adipose Tissue Browning as a Strategy to Combat Obesity. Int J Mol Sci 2020; 21:ijms21176241. [PMID: 32872317 PMCID: PMC7504355 DOI: 10.3390/ijms21176241] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/24/2020] [Accepted: 08/26/2020] [Indexed: 12/25/2022] Open
Abstract
The ongoing obesity pandemic generates a constant need to develop new therapeutic strategies to restore the energy balance. Therefore, the concept of activating brown adipose tissue (BAT) in order to increase energy expenditure has been revived. In mammals, two developmentally distinct types of brown adipocytes exist; the classical or constitutive BAT that arises during embryogenesis, and the beige adipose tissue that is recruited postnatally within white adipose tissue (WAT) in the process called browning. Research of recent years has significantly increased our understanding of the mechanisms involved in BAT activation and WAT browning. They also allowed for the identification of critical molecules and critical steps of both processes and, therefore, many new therapeutic targets. Several non-pharmacological approaches, as well as chemical compounds aiming at the induction of WAT browning and BAT activation, have been tested in vitro as well as in animal models of genetically determined and/or diet-induced obesity. The therapeutic potential of some of these strategies has also been tested in humans. In this review, we summarize present concepts regarding potential therapeutic targets in the process of BAT activation and WAT browning and available strategies aiming at them.
Collapse
Affiliation(s)
- Alina Kuryłowicz
- Department of Human Epigenetics, Mossakowski Medical Research Centre PAS, 02-106 Warsaw, Poland;
- Correspondence: ; Tel.: +48-226086591; Fax: +48-226086410
| | - Monika Puzianowska-Kuźnicka
- Department of Human Epigenetics, Mossakowski Medical Research Centre PAS, 02-106 Warsaw, Poland;
- Department of Geriatrics and Gerontology, Medical Centre of Postgraduate Education, 01-826 Warsaw, Poland
| |
Collapse
|
43
|
Stromal CCL2 Signaling Promotes Mammary Tumor Fibrosis through Recruitment of Myeloid-Lineage Cells. Cancers (Basel) 2020; 12:cancers12082083. [PMID: 32731354 PMCID: PMC7465971 DOI: 10.3390/cancers12082083] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/21/2020] [Accepted: 07/24/2020] [Indexed: 12/18/2022] Open
Abstract
Obesity is correlated with breast tumor desmoplasia, leading to diminished chemotherapy response and disease-free survival. Obesity causes chronic, macrophage-driven inflammation within breast tissue, initiated by chemokine ligand 2 (CCL2) signaling from adipose stromal cells. To understand how CCL2-induced inflammation alters breast tumor pathology, we transplanted oncogenically transformed human breast epithelial cells with breast stromal cells expressing CCL2 or empty vector into murine mammary glands and examined tumor formation and progression with time. As tumors developed, macrophages were rapidly recruited, followed by the emergence of cancer-associated fibroblasts (CAFs) and collagen deposition. Depletion of CD11b + myeloid lineage cells early in tumor formation reduced tumor growth, CAF numbers, and collagen deposition. CCL2 expression within developing tumors also enhanced recruitment of myeloid progenitor cells from the bone marrow into the tumor site. The myeloid progenitor cell population contained elevated numbers of fibrocytes, which exhibited platelet-derived growth factor receptor-alpha (PDGFRα)-dependent colony formation and growth in vitro. Together, these results suggest that chronic inflammation induced by CCL2 significantly enhances tumor growth and promotes the formation of a desmoplastic stroma through early recruitment of macrophages and fibrocytes into the tumor microenvironment. Fibrocytes may be a novel target in the tumor microenvironment to reduce tumor fibrosis and enhance treatment responses for obese breast cancer patients.
Collapse
|
44
|
Maurer S, Harms M, Boucher J. The colorful versatility of adipocytes: white-to-brown transdifferentiation and its therapeutic potential in humans. FEBS J 2020; 288:3628-3646. [PMID: 32621398 DOI: 10.1111/febs.15470] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/17/2020] [Accepted: 06/29/2020] [Indexed: 12/22/2022]
Abstract
Brown and brite adipocytes contribute to energy expenditure through nonshivering thermogenesis. Though these cell types are thought to arise primarily from the de novo differentiation of precursor cells, their abundance is also controlled through the transdifferentiation of mature white adipocytes. Here, we review recent advances in our understanding of the regulation of white-to-brown transdifferentiation, as well as the conversion of brown and brite adipocytes to dormant, white-like fat cells. Converting mature white adipocytes into brite cells or reactivating dormant brown and brite adipocytes has emerged as a strategy to ameliorate human metabolic disorders. We analyze the evidence of learning from mice and how they translate to humans to ultimately scrutinize the relevance of this concept. Moreover, we estimate that converting a small percentage of existing white fat mass in obese subjects into active brite adipocytes could be sufficient to achieve meaningful benefits in metabolism. In conclusion, novel browning agents have to be identified before adipocyte transdifferentiation can be realized as a safe and efficacious therapy.
Collapse
Affiliation(s)
- Stefanie Maurer
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Matthew Harms
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Jeremie Boucher
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.,Lundberg Laboratory for Diabetes Research, University of Gothenburg, Gothenburg, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
45
|
Kaupang Å, Hansen TV. The PPAR Ω Pocket: Renewed Opportunities for Drug Development. PPAR Res 2020; 2020:9657380. [PMID: 32695150 PMCID: PMC7351019 DOI: 10.1155/2020/9657380] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/13/2020] [Indexed: 12/13/2022] Open
Abstract
The past decade of PPARγ research has dramatically improved our understanding of the structural and mechanistic bases for the diverging physiological effects of different classes of PPARγ ligands. The discoveries that lie at the heart of these developments have enabled the design of a new class of PPARγ ligands, capable of isolating central therapeutic effects of PPARγ modulation, while displaying markedly lower toxicities than previous generations of PPARγ ligands. This review examines the emerging framework around the design of these ligands and seeks to unite its principles with the development of new classes of ligands for PPARα and PPARβ/δ. The focus is on the relationships between the binding modes of ligands, their influence on PPAR posttranslational modifications, and gene expression patterns. Specifically, we encourage the design and study of ligands that primarily bind to the Ω pockets of PPARα and PPARβ/δ. In support of this development, we highlight already reported ligands that if studied in the context of this new framework may further our understanding of the gene programs regulated by PPARα and PPARβ/δ. Moreover, recently developed pharmacological tools that can be utilized in the search for ligands with new binding modes are also presented.
Collapse
Affiliation(s)
- Åsmund Kaupang
- Section for Pharmaceutical Chemistry, Department of Pharmacy, University of Oslo, 0316 Oslo, Norway
| | - Trond Vidar Hansen
- Section for Pharmaceutical Chemistry, Department of Pharmacy, University of Oslo, 0316 Oslo, Norway
| |
Collapse
|
46
|
Nikanorova AA, Barashkov NA, Nakhodkin SS, Pshennikova VG, Solovyev AV, Romanov GP, Kuzmina SS, Sazonov NN, Burtseva TE, Odland JØ, Fedorova SA. The Role of Leptin Levels in Adaptation to Cold Climates. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17061854. [PMID: 32178438 PMCID: PMC7143756 DOI: 10.3390/ijerph17061854] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/06/2020] [Accepted: 03/10/2020] [Indexed: 12/21/2022]
Abstract
Currently, adipose tissue is considered an endocrine organ that produces hormone-active substances, including leptin, which can play a key role in thermoregulation processes. Therefore, we performed a meta-analysis to investigate the influence of the climatic environment on leptin levels. A systematic literature search in the databases was carried out on 10 January 2020. Finally, 22 eligible articles were included in the current meta-analysis and a total of 13,320 participants were covered in the final analysis. It was shown that males of the “North” subgroup demonstrated significantly higher levels of leptin (10.02 ng/mL; CI: 7.92–12.13) than males of the “South” subgroup (4.9 ng/mL; CI: 3.71–6.25) (p = 0.0001). On the contrary, in the female group, a similar pattern was not detected (p = 0.91). Apparently, in order to maintain body temperature, higher leptin levels are required. The results of the study indicate that such effects are most pronounced in males and to a smaller extent in females, apparently due to a relatively high initial concentration of leptin in females. The correlation between leptin levels and climatic environment data support the hypothesis of leptin-mediated thermoregulation as an adaptive mechanism to cold climates.
Collapse
Affiliation(s)
- Alena A. Nikanorova
- Laboratory of Molecular Genetics, Yakut Science Centre of Complex Medical Problems, 677010 Yakutsk, Sakha Republic (Yakutia), Russia; (A.A.N.); (S.S.N.); (V.G.P.); (A.V.S.); (G.P.R.); (S.A.F.)
- Laboratory of Molecular Biology, M.K. Ammosov North-Eastern Federal University, Yakutsk, 677000 Sakha Republic (Yakutia), Russia; (S.S.K.); (N.N.S.)
| | - Nikolay A. Barashkov
- Laboratory of Molecular Genetics, Yakut Science Centre of Complex Medical Problems, 677010 Yakutsk, Sakha Republic (Yakutia), Russia; (A.A.N.); (S.S.N.); (V.G.P.); (A.V.S.); (G.P.R.); (S.A.F.)
- Laboratory of Molecular Biology, M.K. Ammosov North-Eastern Federal University, Yakutsk, 677000 Sakha Republic (Yakutia), Russia; (S.S.K.); (N.N.S.)
- Correspondence:
| | - Sergey S. Nakhodkin
- Laboratory of Molecular Genetics, Yakut Science Centre of Complex Medical Problems, 677010 Yakutsk, Sakha Republic (Yakutia), Russia; (A.A.N.); (S.S.N.); (V.G.P.); (A.V.S.); (G.P.R.); (S.A.F.)
- Laboratory of Molecular Biology, M.K. Ammosov North-Eastern Federal University, Yakutsk, 677000 Sakha Republic (Yakutia), Russia; (S.S.K.); (N.N.S.)
| | - Vera G. Pshennikova
- Laboratory of Molecular Genetics, Yakut Science Centre of Complex Medical Problems, 677010 Yakutsk, Sakha Republic (Yakutia), Russia; (A.A.N.); (S.S.N.); (V.G.P.); (A.V.S.); (G.P.R.); (S.A.F.)
- Laboratory of Molecular Biology, M.K. Ammosov North-Eastern Federal University, Yakutsk, 677000 Sakha Republic (Yakutia), Russia; (S.S.K.); (N.N.S.)
| | - Aisen V. Solovyev
- Laboratory of Molecular Genetics, Yakut Science Centre of Complex Medical Problems, 677010 Yakutsk, Sakha Republic (Yakutia), Russia; (A.A.N.); (S.S.N.); (V.G.P.); (A.V.S.); (G.P.R.); (S.A.F.)
- Laboratory of Molecular Biology, M.K. Ammosov North-Eastern Federal University, Yakutsk, 677000 Sakha Republic (Yakutia), Russia; (S.S.K.); (N.N.S.)
- Laboratory of the Human in the Arctic, The Institute for Humanities Research and Indigenous Studies of the North, Federal Research Center “Yakut Science Center of Siberian Branch of Russian Academy of Science”, Yakutsk, 677027 Sakha Republic (Yakutia), Russia
| | - Georgii P. Romanov
- Laboratory of Molecular Genetics, Yakut Science Centre of Complex Medical Problems, 677010 Yakutsk, Sakha Republic (Yakutia), Russia; (A.A.N.); (S.S.N.); (V.G.P.); (A.V.S.); (G.P.R.); (S.A.F.)
- Laboratory of Molecular Biology, M.K. Ammosov North-Eastern Federal University, Yakutsk, 677000 Sakha Republic (Yakutia), Russia; (S.S.K.); (N.N.S.)
| | - Sargylana S. Kuzmina
- Laboratory of Molecular Biology, M.K. Ammosov North-Eastern Federal University, Yakutsk, 677000 Sakha Republic (Yakutia), Russia; (S.S.K.); (N.N.S.)
| | - Nikolay N. Sazonov
- Laboratory of Molecular Biology, M.K. Ammosov North-Eastern Federal University, Yakutsk, 677000 Sakha Republic (Yakutia), Russia; (S.S.K.); (N.N.S.)
| | - Tatyana E. Burtseva
- Department of Pediatrics and Child Surgery, M.K. Ammosov North-Eastern Federal University, Yakutsk, 677000 Sakha Republic (Yakutia), Russia;
- Laboratory of Monitoring Children Health and Medico-environmental Research, Yakut Science Centre of Complex Medical Problems, Yakutsk, 677010 Sakha Republic (Yakutia), Russia
| | - Jon Øyvind Odland
- Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, NTNU Norwegian University of Science and Technology, 7003 Trondheim, Norway;
| | - Sardana A. Fedorova
- Laboratory of Molecular Genetics, Yakut Science Centre of Complex Medical Problems, 677010 Yakutsk, Sakha Republic (Yakutia), Russia; (A.A.N.); (S.S.N.); (V.G.P.); (A.V.S.); (G.P.R.); (S.A.F.)
- Laboratory of Molecular Biology, M.K. Ammosov North-Eastern Federal University, Yakutsk, 677000 Sakha Republic (Yakutia), Russia; (S.S.K.); (N.N.S.)
| |
Collapse
|
47
|
Khim KW, Choi SS, Jang HJ, Lee YH, Lee E, Hyun JM, Eom HJ, Yoon S, Choi JW, Park TE, Nam D, Choi JH. PPM1A Controls Diabetic Gene Programming through Directly Dephosphorylating PPARγ at Ser273. Cells 2020; 9:cells9020343. [PMID: 32024237 PMCID: PMC7072254 DOI: 10.3390/cells9020343] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 01/31/2020] [Indexed: 12/16/2022] Open
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) is a master regulator of adipose tissue biology. In obesity, phosphorylation of PPARγ at Ser273 (pSer273) by cyclin-dependent kinase 5 (CDK5)/extracellular signal-regulated kinase (ERK) orchestrates diabetic gene reprogramming via dysregulation of specific gene expression. Although many recent studies have focused on the development of non-classical agonist drugs that inhibit the phosphorylation of PPARγ at Ser273, the molecular mechanism of PPARγ dephosphorylation at Ser273 is not well characterized. Here, we report that protein phosphatase Mg2+/Mn2+-dependent 1A (PPM1A) is a novel PPARγ phosphatase that directly dephosphorylates Ser273 and restores diabetic gene expression which is dysregulated by pSer273. The expression of PPM1A significantly decreases in two models of insulin resistance: diet-induced obese (DIO) mice and db/db mice, in which it negatively correlates with pSer273. Transcriptomic analysis using microarray and genotype-tissue expression (GTEx) data in humans shows positive correlations between PPM1A and most of the genes that are dysregulated by pSer273. These findings suggest that PPM1A dephosphorylates PPARγ at Ser273 and represents a potential target for the treatment of obesity-linked metabolic disorders.
Collapse
|
48
|
Structural Basis for the Regulation of PPARγ Activity by Imatinib. Molecules 2019; 24:molecules24193562. [PMID: 31581474 PMCID: PMC6803859 DOI: 10.3390/molecules24193562] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/27/2019] [Accepted: 09/30/2019] [Indexed: 12/17/2022] Open
Abstract
Imatinib is an effective anticancer drug for the treatment of leukemia. Interestingly, when an FDA-approved drug library was tested for agents that block peroxisome proliferator-activated receptor γ (PPARγ) phosphorylation at Ser245 to evaluate possibilities of antidiabetic drug repositioning, imatinib was determined as a PPARγ antagonist ligand. However, it is not well understood how imatinib binds to PPARγ or would improve insulin sensitivity without classical agonism. Here, we report the crystal structure of the PPARγ R288A mutant in complex with imatinib. Imatinib bound to Arm2 and Arm3 regions in the ligand-binding domain (LBD) of PPARγ, of which the Arm3 region is closely related to the inhibition of PPARγ phosphorylation at Ser245. The binding of imatinib in LBD induced a stable conformation of helix H2′ and the Ω loop compared with the ligand-free state. In contrast, imatinib does not interact with Tyr473 on PPARγ helix H12, which is important for the classical agonism associated with side effects. Our study provides new structural insights into the PPARγ regulation by imatinib and may contribute to the development of new antidiabetic drugs targeting PPARγ while minimizing known side effects.
Collapse
|
49
|
Li J, Cha R, Luo H, Hao W, Zhang Y, Jiang X. Nanomaterials for the theranostics of obesity. Biomaterials 2019; 223:119474. [PMID: 31536920 DOI: 10.1016/j.biomaterials.2019.119474] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 09/01/2019] [Accepted: 09/03/2019] [Indexed: 02/06/2023]
Abstract
As a chronic and lifelong disease, obesity not only significant impairs health but also dramatically shortens life span (at least 10 years). Obesity requires a life-long effort for the successful treatment because a number of abnormalities would appear in the development of obesity. Nanomaterials possess large specific surface area, strong absorptivity, and high bioavailability, especially the good targeting properties and adjustable release rate, which would benefit the diagnosis and treatment of obesity and obesity-related metabolic diseases. Herein, we discussed the therapy and diagnosis of obesity and obesity-related metabolic diseases by using nanomaterials. Therapies of obesity with nanomaterials include improving intestinal health and reducing energy intake, targeting and treating functional cell abnormalities, regulating redox homeostasis, and removing free lipoprotein in blood. Diagnosis of obesity-related metabolic diseases would benefit the therapy of these diseases. The development of nanomaterials will promote the diagnosis and therapy of obesity and obesity-related metabolic diseases.
Collapse
Affiliation(s)
- Juanjuan Li
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, No. 11 Zhongguancun Beiyitiao, Beijing, 100190, PR China
| | - Ruitao Cha
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, No. 11 Zhongguancun Beiyitiao, Beijing, 100190, PR China.
| | - Huize Luo
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, No. 11 Zhongguancun Beiyitiao, Beijing, 100190, PR China
| | - Wenshuai Hao
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, No. 11 Zhongguancun Beiyitiao, Beijing, 100190, PR China
| | - Yan Zhang
- Department of Cardiac Surgery, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100032, PR China.
| | - Xingyu Jiang
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, No. 11 Zhongguancun Beiyitiao, Beijing, 100190, PR China; Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen, Guangdong, 518055, PR China; University of Chinese Academy of Sciences, 19 A Yuquan Road, Shijingshan District, Beijing, 100049, PR China.
| |
Collapse
|
50
|
Fang D, Shi X, Lu T, Ruan H, Gao Y. The glycoprotein follistatin-like 1 promotes brown adipose thermogenesis. Metabolism 2019; 98:16-26. [PMID: 31132382 DOI: 10.1016/j.metabol.2019.05.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 05/10/2019] [Accepted: 05/21/2019] [Indexed: 10/26/2022]
Abstract
OBJECTIVES The thermogenic brown adipose tissue (BAT) has been proposed as a potential target to prevent or treat obesity and related metabolic diseases. BAT secretes adipokines to regulate the thermogenic program in an autocrine or paracrine manner. Follistatin-like 1 (FSTL1), a glycoprotein involved in adipogenesis and obesity, however, the function of FSTL1 in BAT thermogenesis and in the regulation of systemic energy homeostasis are not fully understood. METHODS Whole-body ablation Fstl1 heterozygous mice (Fstl1+/-) and its littermates control were injected with CL316,243 to assess energy balance. A series of FSTL1 overexpression and knockdown experiments were carried out to evaluate its function in regulating thermogenic gene expression in brown adipocytes. RESULTS FSTL1 expression was induced upon BAT activation during cold challenge or β3-adrenergic activation. FSTL1 haploinsufficiency in mice led to reduced thermogenic gene expression, impaired BAT recruitment, and decreased heat production. FSTL1 cell-autonomously promoted the β3-adrenergic signaling, which was required to upregulate PPARγ and UCP1 in brown adipocytes. Furthermore, only glycosylated FSTL1 could be secreted from brown adipocytes to induce the β3-adrenergic activation. CONCLUSIONS Our results suggest FSTL1 as a novel stimulator of the β-adrenergic signaling and BAT thermogenesis.
Collapse
Affiliation(s)
- Dongliang Fang
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Human Anatomy, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Xinyi Shi
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Human Anatomy, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Tao Lu
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Human Anatomy, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Haibin Ruan
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Yan Gao
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Human Anatomy, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China.
| |
Collapse
|