1
|
Kiani P, Khodadadi ES, Nikdasti A, Yarahmadi S, Gheibi M, Yousefi Z, Ehtiati S, Yahyazadeh S, Shafiee SM, Taghizadeh M, Igder S, Khatami SH, Karima S, Vakili O, Pourfarzam M. Autophagy and the peroxisome proliferator-activated receptor signaling pathway: A molecular ballet in lipid metabolism and homeostasis. Mol Cell Biochem 2025; 480:3477-3499. [PMID: 39891864 DOI: 10.1007/s11010-025-05207-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/04/2025] [Indexed: 02/03/2025]
Abstract
Lipids, which are indispensable for cellular architecture and energy storage, predominantly consist of triglycerides (TGs), phospholipids, cholesterol, and their derivatives. These hydrophobic entities are housed within dynamic lipid droplets (LDs), which expand and contract in response to nutrient availability. Historically perceived as a cellular waste disposal mechanism, autophagy has now been recognized as a crucial regulator of metabolism. Within this framework, lipophagy, the selective degradation of LDs, plays a fundamental role in maintaining lipid homeostasis. Dysregulated lipid metabolism and autophagy are frequently associated with metabolic disorders such as obesity and atherosclerosis. In this context, peroxisome proliferator-activated receptors (PPARs), particularly PPAR-γ, serve as intracellular lipid sensors and master regulators of gene expression. Their regulatory influence extends to both autophagy and lipid metabolism, indicating a complex interplay between these processes. This review explores the hypothesis that PPARs may directly modulate autophagy within the realm of lipid metabolism, thereby contributing to the pathogenesis of metabolic diseases. By elucidating the underlying molecular mechanisms, we aim to provide a comprehensive understanding of the intricate regulatory network that connects PPARs, autophagy, and lipid homeostasis. The crosstalk between PPARs and other signaling pathways underscores the complexity of their regulatory functions and the potential for therapeutic interventions targeting these pathways. The intricate relationships among PPARs, autophagy, and lipid metabolism represent a pivotal area of research with significant implications for understanding and treating metabolic disorders.
Collapse
Affiliation(s)
- Pouria Kiani
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elaheh Sadat Khodadadi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122, Padova, Italy
| | - Ali Nikdasti
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020, Legnaro, Padova, Italy
| | - Sahar Yarahmadi
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mobina Gheibi
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zeynab Yousefi
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sajad Ehtiati
- Student Research Committee, Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sheida Yahyazadeh
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sayed Mohammad Shafiee
- Autophagy Research Center, Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Motahareh Taghizadeh
- Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Somayeh Igder
- Department of Clinical Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyyed Hossein Khatami
- Student Research Committee, Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Saeed Karima
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran.
| | - Omid Vakili
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Morteza Pourfarzam
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
2
|
Warda F, Batch J, Graham L, Haas MJ, Mooradian AD. D-allulose enhances lipid oxidation in HepG2 cells via peroxisome proliferator-activated receptor α (PPARα). Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159599. [PMID: 39884381 DOI: 10.1016/j.bbalip.2025.159599] [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: 03/28/2024] [Revised: 12/31/2024] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
Lipid accumulation in hepatocytes in non-alcoholic steatohepatitis (NASH) is attributed partly to loss of insulin-responsiveness and/or an increased pro-inflammatory state. Since the rare sugar D-allulose has insulin mimetic and anti-inflammatory properties, its effects on lipid accumulation in liver-derived cells was tested. In HepG2 cells exposed to 200 μM oleic acid for 72 h, D-allulose treatment decreased intracellular lipid accumulation with an IC50 = 0.45 ± 0.07 mM. A similar effect was observed in cells treated with 10 μM gemfibrozil. D-allulose and gemfibrozil treatment increased oleic acid β-oxidation. Both D-allulose and gemfibrozil increased peroxisome proliferator-activated receptor α (PPARα) expression (two-fold) relative to control cells, while retinoid X receptor was unchanged. D-allulose and gemfibrozil increased PPARα-dependent genes including those involved in fatty acid β-oxidation (acyl-coenzyme A oxidase 1, long-chain-fatty-acid-coenzyme A ligase 5, and carnitine palmitoyltransferase 1 A). D-allulose and gemfibrozil also increased PPARα reporter gene expression and phosphorylation (Serine 12) which were both inhibited by the mitogen-activated protein (MAP) kinase inhibitor PD098059. Other MAP kinase inhibitors, including SB203580, SP600125, and BIX10289 had no effect on reporter gene expression. Oleic acid treatment, but not D-allulose or gemfibrozil, decreased sterol response element binding protein 1 and sterol response element binding protein 2 expression relative to cells not exposed to oleic acid, while peroxisome proliferator-activated receptor γ expression did not change. These results indicate that D-alluose mimics gemfibrozil effects on lipid content in HepG2 cells by promoting fatty acid β-oxidation via PPARα.
Collapse
Affiliation(s)
- Firas Warda
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida Jacksonville College of Medicine, Jacksonville, FL 32209, United States of America
| | - Jennifer Batch
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida Jacksonville College of Medicine, Jacksonville, FL 32209, United States of America
| | - Lauren Graham
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida Jacksonville College of Medicine, Jacksonville, FL 32209, United States of America
| | - Michael J Haas
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida Jacksonville College of Medicine, Jacksonville, FL 32209, United States of America.
| | - Arshag D Mooradian
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida Jacksonville College of Medicine, Jacksonville, FL 32209, United States of America
| |
Collapse
|
3
|
Kim A, Jung S, Kim Y, Jung J, Lee S, Lee H, Kim MJ, Park JY, Hwang EM, Lee J. Novel function of TREK-1 in regulating adipocyte differentiation and lipid accumulation. Cell Death Dis 2025; 16:164. [PMID: 40057491 PMCID: PMC11890776 DOI: 10.1038/s41419-025-07478-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 02/07/2025] [Accepted: 02/24/2025] [Indexed: 05/13/2025]
Abstract
K2P (two-pore domain potassium) channels, a diversified class of K+-selective ion channels, have been found to affect a wide range of physiological processes in the body. Despite their established significance in regulating proliferation and differentiation in multiple cell types, K2P channels' specific role in adipogenic differentiation (adipogenesis) remains poorly understood. In this study, we investigated the engagement of K2P channels, specifically KCNK2 (also known as TREK-1), in adipogenesis using primary cultured adipocytes and TREK-1 knockout (KO) mice. Our findings showed that TREK-1 expression in adipocytes decreases substantially during adipogenesis. This typically causes an increased Ca2+ influx and alters the electrical potential of the cell membrane in 3T3-L1 cell lines. Furthermore, we observed an increase in differentiation and lipid accumulation in both 3T3-L1 cell lines and primary cultured adipocytes when the TREK-1 activity was blocked with Spadin, the specific inhibitors, and TREK-1 shRNA. Finally, our findings revealed that mice lacking TREK-1 gained more fat mass and had worse glucose tolerance when fed a high-fat diet (HFD) compared to the wild-type controls. The findings demonstrate that increase of the membrane potential at adipocytes through the downregulation of TREK-1 can influence the progression of adipogenesis.
Collapse
Affiliation(s)
- Ajung Kim
- Food Functionality Research Division, Korea Food Research Institute, Wanju, 55365, South Korea
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, 02792, South Korea
| | - Seoyeong Jung
- Food Functionality Research Division, Korea Food Research Institute, Wanju, 55365, South Korea
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul, 02841, South Korea
| | - Yongeun Kim
- Food Functionality Research Division, Korea Food Research Institute, Wanju, 55365, South Korea
| | - Jonghoon Jung
- Food Functionality Research Division, Korea Food Research Institute, Wanju, 55365, South Korea
| | - Soomin Lee
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, 02792, South Korea
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul, 02841, South Korea
| | - Hojin Lee
- Food Functionality Research Division, Korea Food Research Institute, Wanju, 55365, South Korea
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul, 02841, South Korea
| | - Min Jung Kim
- Food Functionality Research Division, Korea Food Research Institute, Wanju, 55365, South Korea
| | - Jae-Yong Park
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul, 02841, South Korea
| | - Eun Mi Hwang
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, 02792, South Korea.
| | - Jaekwang Lee
- Food Functionality Research Division, Korea Food Research Institute, Wanju, 55365, South Korea.
| |
Collapse
|
4
|
Fougerat A, Bruse J, Polizzi A, Montagner A, Guillou H, Wahli W. Lipid sensing by PPARα: Role in controlling hepatocyte gene regulatory networks and the metabolic response to fasting. Prog Lipid Res 2024; 96:101303. [PMID: 39521352 DOI: 10.1016/j.plipres.2024.101303] [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: 07/17/2024] [Revised: 10/18/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Peroxisome proliferator-activated receptors (PPARs) constitute a small family of three nuclear receptors that act as lipid sensors, and thereby regulate the transcription of genes having key roles in hepatic and whole-body energy homeostasis, and in other processes (e.g., inflammation), which have far-reaching health consequences. Peroxisome proliferator-activated receptor isotype α (PPARα) is expressed in oxidative tissues, particularly in the liver, carrying out critical functions during the adaptive fasting response. Advanced omics technologies have provided insight into the vast complexity of the regulation of PPAR expression and activity, as well as their downstream effects on the physiology of the liver and its associated metabolic organs. Here, we provide an overview of the gene regulatory networks controlled by PPARα in the liver in response to fasting. We discuss impacts on liver metabolism, the systemic repercussions and benefits of PPARα-regulated ketogenesis and production of fibroblast growth factor 21 (FGF21), a fasting- and stress-inducible metabolic hormone. We also highlight current challenges in using novel methods to further improve our knowledge of PPARα in health and disease.
Collapse
Affiliation(s)
- Anne Fougerat
- Toxalim (Research Centre in Toxicology), INRAE, ENVT, INP-Purpan, UPS, Toulouse University, Toulouse, France.
| | - Justine Bruse
- Toxalim (Research Centre in Toxicology), INRAE, ENVT, INP-Purpan, UPS, Toulouse University, Toulouse, France
| | - Arnaud Polizzi
- Toxalim (Research Centre in Toxicology), INRAE, ENVT, INP-Purpan, UPS, Toulouse University, Toulouse, France
| | - Alexandra Montagner
- Institute of Metabolic and Cardiovascular Diseases (I2MC), INSERM UMR1297, Toulouse III University, University Paul Sabatier (UPS), Toulouse, France
| | - Hervé Guillou
- Toxalim (Research Centre in Toxicology), INRAE, ENVT, INP-Purpan, UPS, Toulouse University, Toulouse, France
| | - Walter Wahli
- Toxalim (Research Centre in Toxicology), INRAE, ENVT, INP-Purpan, UPS, Toulouse University, Toulouse, France; Center for Integrative Genomics, University of Lausanne, CH-1015 Lausanne, Switzerland.
| |
Collapse
|
5
|
Leya M, Jeong H, Yang D, Ton Nu Bao TH, Pandeya PR, Oh SI, Roh YS, Kim JW, Kim B. Hepatocyte-Specific Casein Kinase 1 Epsilon Ablation Ameliorates Metabolic Dysfunction-Associated Steatohepatitis by Up-Regulating Tumor Necrosis Factor Receptor-Associated Factor 3 in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:2106-2127. [PMID: 39179201 DOI: 10.1016/j.ajpath.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/09/2024] [Accepted: 08/02/2024] [Indexed: 08/26/2024]
Abstract
Casein kinase 1 epsilon (CK1ε), a member of the serine/threonine protein kinase family, phosphorylates a broad range of substrates. However, its role in the development of chronic liver diseases remains elusive. This study aimed to investigate the role of CK1ε in the development and progression of metabolic dysfunction-associated steatohepatitis (MASH). Hepatocyte-specific CK1ε knockout (CK1εΔHEP) mice were generated by crossbreeding mice with floxed CK1ε alleles (CK1εfl/fl) and Cre-expressing albumin mice. Mice were fed either a Western diet (WD) or a methionine- and choline-deficient diet to induce MASH. CK1εΔHEP was associated with a decreased severity of WD- or methionine- and choline-deficient diet-induced MASH, as confirmed by reduced incidence of hepatic lesions and significantly lower levels of alanine aminotransferase, aspartate aminotransferase, and proinflammatory cytokine tumor necrosis factor (TNF)-α. CK1εΔHEP WD-fed mice exhibited significant amelioration of total cholesterol, triglycerides, and de novo lipogenic genes, indicating that CK1ε could influence lipid metabolism. CK1εΔHEP WD-fed mice showed significantly down-regulated TNF receptor-associated factor (TRAF) 3, phosphorylated (p) transforming growth factor-β-activated kinase 1, p-TRAF-associated NF-κB activator (TANK)-binding kinase 1 (TBK1), and p-AKT levels, thereby affecting downstream mitogen-activated protein kinase signaling, indicating a potential mechanism for the observed rescue. Finally, pharmacologic inhibition of CK1ε with PF670462 improved palmitic acid-induced steatohepatitis in vitro and attenuated WD-induced metabolic profile in vivo. In conclusion, CK1ε up-regulates TNF receptor-associated factor 3, which, in turn, causes transforming growth factor-β-activated kinase 1-dependent signaling, amplifies downstream mitogen-activated protein kinase signaling, modifies p-c-Jun levels, and exacerbates inflammation, all of which are factors in WD-induced metabolic dysfunction-associated steatotic liver disease.
Collapse
Affiliation(s)
- Mwense Leya
- Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan-si, Republic of Korea; School of Veterinary Medicine, University of Namibia, Windhoek, Namibia
| | - Hyuneui Jeong
- Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan-si, Republic of Korea
| | - Daram Yang
- Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan-si, Republic of Korea
| | - Tien Huyen Ton Nu Bao
- Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan-si, Republic of Korea
| | - Prakash Raj Pandeya
- Department of Animal and Food Sciences, University of Kentucky, Lexington, Kentucky
| | - Sang-Ik Oh
- Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan-si, Republic of Korea
| | - Yoon-Seok Roh
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju-si, Republic of Korea
| | - Jong-Won Kim
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania.
| | - Bumseok Kim
- Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan-si, Republic of Korea.
| |
Collapse
|
6
|
Kim S, Na GH, Yim DJ, Liu CF, Lin TH, Shih TW, Pan TM, Lee CL, Koo YK. Lactobacillus paracasei subsp. paracasei NTU 101 prevents obesity by regulating AMPK pathways and gut microbiota in obese rat. Biochem Biophys Res Commun 2024; 731:150279. [PMID: 39018972 DOI: 10.1016/j.bbrc.2024.150279] [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: 03/27/2024] [Revised: 06/10/2024] [Accepted: 06/17/2024] [Indexed: 07/19/2024]
Abstract
This study assessed the anti-obesity effects of Lactobacillus paracasei subsp. paracasei NTU 101 (NTU 101) both in vitro and in vivo. Initially, the cytotoxicity and lipid accumulation inhibitory effects of NTU 101 on 3T3-L1 cells were evaluated using the MTT assay and oil red O assay, respectively. Subsequently, the anti-obesity effects of NTU 101 were investigated in high-fat diet-induced obese rat. Moreover, western blotting was performed to measure the obesity-related protein expression of PPARα, PPARβ, PPARγ, C/EBPα, C/EBPβ, ATGL, p-p38 MAPK, p-ERK1/2, p-AMPK and CPT-1 in both 3T3-L1 adipocytes and adipose and liver tissues. Treatment with 16 × 108 CFU/mL NTU 101 reduced lipid accumulation in 3T3-L1 adipocytes by more than 50 %. Oral administration of NTU 101 significantly attenuated body weight gain, as well as adipose tissue weight. NTU 101 administration enhanced fatty acid oxidation increasing expression levels of PPARα, CPT-1, and p-AMPK proteins in liver tissue, while simultaneously inhibited adipogenesis by reducing PPARγ and C/EBPα proteins in adipose tissue. Furthermore, NTU 101 supplementation positively modulated the composition of gut microbiota, notably increasing the abundance of Akkermansiaceae. This present study suggests that NTU 101 exerts anti-obesity effects by regulating gut microbiota, fatty acid oxidation, lipolysis and adipogenesis.
Collapse
Affiliation(s)
- SukJin Kim
- Department of R&I Center, COSMAXBIO, Seongnam, 13486, South Korea
| | - Gwi Hwan Na
- Department of R&I Center, COSMAXBIO, Seongnam, 13486, South Korea
| | - Dong Joon Yim
- Department of R&I Center, COSMAXBIO, Seongnam, 13486, South Korea
| | - Chin-Feng Liu
- Continuing Education Program of Food Biotechnology Applications, National Taitung University, Taitung, Taiwan, ROC
| | - Tse-Han Lin
- Department of Life Science, National Taitung University, 369, Sec. 2, University Rd., Taitung, 95092, Taitung, Taiwan, ROC
| | | | - Tzu-Ming Pan
- SunWay Biotech Co. LTD., Taipei, Taiwan, ROC; Department of Biochemical Science and Technology, National Taiwan University, Taipei, Taiwan, ROC
| | - Chun-Lin Lee
- Continuing Education Program of Food Biotechnology Applications, National Taitung University, Taitung, Taiwan, ROC.
| | - Yean Kyoung Koo
- Department of R&I Center, COSMAXBIO, Seongnam, 13486, South Korea.
| |
Collapse
|
7
|
Cohen ED, Roethlin K, Yee M, Woeller CF, Brookes PS, Porter GA, O'Reilly MA. PPARγ drives mitochondrial stress signaling and the loss of atrial cardiomyocytes in newborn mice exposed to hyperoxia. Redox Biol 2024; 76:103351. [PMID: 39276392 PMCID: PMC11417530 DOI: 10.1016/j.redox.2024.103351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/22/2024] [Accepted: 09/09/2024] [Indexed: 09/17/2024] Open
Abstract
Diastolic dysfunction is increasingly common in preterm infants exposed to supplemental oxygen (hyperoxia). Previous studies in neonatal mice showed hyperoxia suppresses fatty acid synthesis genes required for proliferation and survival of atrial cardiomyocytes. The loss of atrial cardiomyocytes creates a hypoplastic left atrium that inappropriately fills the left ventricle during diastole. Here, we show that hyperoxia stimulates adenosine monophosphate-activated kinase (AMPK) and peroxisome proliferator activated receptor-gamma (PPARγ) signaling in atrial cardiomyocytes. While both pathways can regulate lipid homeostasis, PPARγ was the primary pathway by which hyperoxia inhibits fatty acid gene expression and inhibits proliferation of mouse atrial HL-1 cells. It also enhanced the toxicity of hyperoxia by increasing expression of activating transcription factor (ATF) 5 and other mitochondrial stress response genes. Silencing PPARγ signaling restored proliferation and survival of HL-1 cells as well as atrial cardiomyocytes in neonatal mice exposed to hyperoxia. Our findings reveal PPARγ enhances the toxicity of hyperoxia on atrial cardiomyocytes, thus suggesting inhibitors of PPARγ signaling may prevent diastolic dysfunction in preterm infants.
Collapse
Affiliation(s)
- E David Cohen
- Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, Rochester, NY 14642, USA.
| | - Kyle Roethlin
- Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, Rochester, NY 14642, USA
| | - Min Yee
- Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, Rochester, NY 14642, USA
| | - Collynn F Woeller
- Department of Ophthalmology, School of Medicine and Dentistry, The University of Rochester, Rochester, NY 14642, USA
| | - Paul S Brookes
- Department of Anesthesiology, School of Medicine and Dentistry, The University of Rochester, Rochester, NY 14642, USA
| | - George A Porter
- Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, Rochester, NY 14642, USA
| | - Michael A O'Reilly
- Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, Rochester, NY 14642, USA.
| |
Collapse
|
8
|
Kumar A, Laborit Labrada B, Lavallée-Bourget MH, Forest MP, Schwab M, Bellmann K, Houde V, Beauchemin N, Laplante M, Marette A. Regulation of PPARγ2 Stability and Activity by SHP-1. Mol Cell Biol 2024; 44:261-272. [PMID: 38828991 PMCID: PMC11253886 DOI: 10.1080/10985549.2024.2354959] [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: 07/03/2023] [Accepted: 04/23/2024] [Indexed: 06/05/2024] Open
Abstract
The protein tyrosine phosphatase Src homology region 2 domain-containing phosphatase-1 (SHP-1) plays an important role in modulating glucose and lipid homeostasis. We previously suggested a potential role of SHP-1 in the regulation of peroxisome proliferator-activated receptor γ2 (PPARγ2) expression and activity but the mechanisms were unexplored. PPARγ2 is the master regulator of adipogenesis, but how its activity is regulated by tyrosine phosphorylation is largely unknown. Here, we found that SHP-1 binds to PPARγ2 primarily via its N-terminal SH2-domain. We confirmed the phosphorylation of PPARγ2 on tyrosine-residue 78 (Y78), which was reduced by SHP-1 in vitro resulting in decreased PPARγ2 stability. Loss of SHP-1 led to elevated, agonist-induced expression of the classical PPARγ2 targets FABP4 and CD36, concomitant with increased lipid content in cells expressing PPARγ2, an effect blunted by abrogation of PPARγ2 phosphorylation. Collectively, we discovered that SHP-1 affects the stability of PPARγ2 through dephosphorylation thereby influencing adipogenesis.
Collapse
Affiliation(s)
- Amit Kumar
- Centre de recherche de l‘Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Beisy Laborit Labrada
- Centre de recherche de l‘Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Marie-Hélène Lavallée-Bourget
- Centre de recherche de l‘Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Marie-Pier Forest
- Centre de recherche de l‘Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Michael Schwab
- Centre de recherche de l‘Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Kerstin Bellmann
- Centre de recherche de l‘Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Vanessa Houde
- Centre de recherche de l‘Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Nicole Beauchemin
- Rosalind and Morris Goodman Cancer Research Centre, Departments of Oncology, Medicine and Biochemistry, McGill University, Montreal, QC, Canada
| | - Mathieu Laplante
- Centre de recherche de l‘Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, QC, Canada
- Centre de Recherche sur le Cancer, l’Université Laval, Québec, QC, Canada
| | - André Marette
- Centre de recherche de l‘Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, QC, Canada
- Institute of Nutrition and Functional Foods, Laval University, Québec, QC, Canada
| |
Collapse
|
9
|
Desmet SJ, Thommis J, Vanderhaeghen T, Vandenboorn EMF, Clarisse D, Li Y, Timmermans S, Fijalkowska D, Ratman D, Van Hamme E, De Cauwer L, Staels B, Brunsveld L, Peelman F, Libert C, Tavernier J, De Bosscher K. Crosstalk interactions between transcription factors ERRα and PPARα assist PPARα-mediated gene expression. Mol Metab 2024; 84:101938. [PMID: 38631478 PMCID: PMC11059514 DOI: 10.1016/j.molmet.2024.101938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/10/2024] [Accepted: 04/09/2024] [Indexed: 04/19/2024] Open
Abstract
OBJECTIVE The peroxisome proliferator-activated receptor α (PPARα) is a transcription factor driving target genes involved in fatty acid β-oxidation. To what extent various PPARα interacting proteins may assist its function as a transcription factor is incompletely understood. An ORFeome-wide unbiased mammalian protein-protein interaction trap (MAPPIT) using PPARα as bait revealed a PPARα-ligand-dependent interaction with the orphan nuclear receptor estrogen-related receptor α (ERRα). The goal of this study was to characterize the nature of the interaction in depth and to explore whether it was of physiological relevance. METHODS We used orthogonal protein-protein interaction assays and pharmacological inhibitors of ERRα in various systems to confirm a functional interaction and study the impact of crosstalk mechanisms. To characterize the interaction surfaces and contact points we applied a random mutagenesis screen and structural overlays. We pinpointed the extent of reciprocal ligand effects of both nuclear receptors via coregulator peptide recruitment assays. On PPARα targets revealed from a genome-wide transcriptome analysis, we performed an ERRα chromatin immunoprecipitation analysis on both fast and fed mouse livers. RESULTS Random mutagenesis scanning of PPARα's ligand-binding domain and coregulator profiling experiments supported the involvement of (a) bridging coregulator(s), while recapitulation of the interaction in vitro indicated the possibility of a trimeric interaction with RXRα. The PPARα·ERRα interaction depends on 3 C-terminal residues within helix 12 of ERRα and is strengthened by both PGC1α and serum deprivation. Pharmacological inhibition of ERRα decreased the interaction of ERRα to ligand-activated PPARα and revealed a transcriptome in line with enhanced mRNA expression of prototypical PPARα target genes, suggesting a role for ERRα as a transcriptional repressor. Strikingly, on other PPARα targets, including the isolated PDK4 enhancer, ERRα behaved oppositely. Chromatin immunoprecipitation analyses demonstrate a PPARα ligand-dependent ERRα recruitment onto chromatin at PPARα-binding regions, which is lost following ERRα inhibition in fed mouse livers. CONCLUSIONS Our data support the coexistence of multiple layers of transcriptional crosstalk mechanisms between PPARα and ERRα, which may serve to finetune the activity of PPARα as a nutrient-sensing transcription factor.
Collapse
Affiliation(s)
- Sofie J Desmet
- VIB Center for Medical Biotechnology, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Jonathan Thommis
- VIB Center for Medical Biotechnology, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Tineke Vanderhaeghen
- VIB Center for Inflammation Research, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Edmee M F Vandenboorn
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, 5612AZ Eindhoven, the Netherlands
| | - Dorien Clarisse
- VIB Center for Medical Biotechnology, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Yunkun Li
- VIB Center for Medical Biotechnology, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Steven Timmermans
- VIB Center for Inflammation Research, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Daria Fijalkowska
- VIB Center for Medical Biotechnology, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Dariusz Ratman
- VIB Center for Medical Biotechnology, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | | | - Lode De Cauwer
- VIB Center for Medical Biotechnology, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Bart Staels
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Luc Brunsveld
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, 5612AZ Eindhoven, the Netherlands
| | - Frank Peelman
- VIB Center for Medical Biotechnology, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Claude Libert
- VIB Center for Inflammation Research, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Jan Tavernier
- VIB Center for Medical Biotechnology, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Karolien De Bosscher
- VIB Center for Medical Biotechnology, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium.
| |
Collapse
|
10
|
Meng X, Wang L, Du YC, Cheng D, Zeng T. PPARβ/δ as a promising molecular drug target for liver diseases: A focused review. Clin Res Hepatol Gastroenterol 2024; 48:102343. [PMID: 38641250 DOI: 10.1016/j.clinre.2024.102343] [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: 12/27/2023] [Revised: 04/03/2024] [Accepted: 04/17/2024] [Indexed: 04/21/2024]
Abstract
Various liver diseases pose great threats to humans. Although the etiologies of these liver diseases are quite diverse, they share similar pathologic phenotypes and molecular mechanisms such as oxidative stress, lipid and glucose metabolism disturbance, hepatic Kupffer cell (KC) proinflammatory polarization and inflammation, insulin resistance, and hepatic stellate cell (HSC) activation and proliferation. Peroxisome proliferator-activated receptor β/δ (PPARβ/δ) is expressed in various types of liver cells with relatively higher expression in KCs and HSCs. Accumulating evidence has revealed the versatile functions of PPARβ/δ such as controlling lipid homeostasis, inhibiting inflammation, regulating glucose metabolism, and restoring insulin sensitivity, suggesting that PPARβ/δ may serve as a potential molecular drug target for various liver diseases. This article aims to provide a concise review of the structure, expression pattern and biological functions of PPARβ/δ in the liver and its roles in various liver diseases, and to discuss potential future research perspectives.
Collapse
Affiliation(s)
- Xin Meng
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Lin Wang
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yan-Chao Du
- Jinan Institute for Product Quality Inspection, Jinan, Shandong 250102, China
| | - Dong Cheng
- Department of Health Test and Detection, Shandong Center for Disease Control and Prevention, Jinan, Shandong 250014, China.
| | - Tao Zeng
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
11
|
Wang Y, Ren J, Ren S. Larsucosterol: endogenous epigenetic regulator for treating chronic and acute liver diseases. Am J Physiol Endocrinol Metab 2024; 326:E577-E587. [PMID: 38381400 PMCID: PMC11376820 DOI: 10.1152/ajpendo.00406.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/15/2024] [Accepted: 02/15/2024] [Indexed: 02/22/2024]
Abstract
Larsucosterol, a potent endogenous epigenetic regulator, has been reported to play a significant role in lipid metabolism, inflammatory responses, and cell survival. The administration of larsucosterol has demonstrated a reduction in lipid accumulation within hepatocytes and the attenuation of inflammatory responses induced by lipopolysaccharide (LPS) and TNFα in macrophages, alleviating LPS- and acetaminophen (ATMP)-induced multiple organ injury, and decreasing mortalities in animal models. Results from phase 1 and 2 clinical trials have shown that larsucosterol has potential as a biomedicine for the treatment of acute and chronic liver diseases. Recent evidence suggests that larsucosterol is a promising candidate for treating alcohol-associated hepatitis with positive results from a phase 2a clinical trial, and for metabolic dysfunction-associated steatohepatitis (MASH) from a phase 1b clinical trial. In this review, we present a culmination of our recent research efforts spanning two decades. We summarize the discovery, physiological and pharmacological mechanisms, and clinical applications of larsucosterol. Furthermore, we elucidate the pathophysiological pathways of metabolic dysfunction-associated steatotic liver diseases (MASLD), metabolic dysfunction-associated steatohepatitis (MASH), and acute liver injuries. A central focus of the review is the exploration of the therapeutic potential of larsucosterol in treating life-threatening conditions, including acetaminophen overdose, endotoxin shock, MASLD, MASH, hepatectomy, and alcoholic hepatitis.
Collapse
Affiliation(s)
- Yaping Wang
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
- McGuire Veterans Affairs Medical Center, Richmond, Virginia, United States
| | - Jenna Ren
- Department of Pharmacology, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Shunlin Ren
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
- McGuire Veterans Affairs Medical Center, Richmond, Virginia, United States
| |
Collapse
|
12
|
Sharma S, Sharma D, Dhobi M, Wang D, Tewari D. An insight to treat cardiovascular diseases through phytochemicals targeting PPAR-α. Mol Cell Biochem 2024; 479:707-732. [PMID: 37171724 DOI: 10.1007/s11010-023-04755-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 04/28/2023] [Indexed: 05/13/2023]
Abstract
Peroxisome proliferator-activated receptor-α (PPAR-α) belonging to the nuclear hormone receptor superfamily is a promising target for CVDs which mechanistically improves the production of high-density lipid as well as inhibit vascular smooth muscle cell proliferation. PPAR-α mainly interferes with adenosine monophosphate-activated protein kinase, transforming growth factor-β-activated kinase, and nuclear factor-κB pathways to protect against cardiac complications. Natural products/extracts could serve as a potential therapeutic strategy in CVDs for targeting PPAR-α with broad safety margins. In recent years, the understanding of naturally derived PPAR-α agonists has considerably improved; however, the information is scattered. In vitro and in vivo studies on acacetin, apigenin, arjunolic acid, astaxanthin, berberine, resveratrol, vaticanol C, hispidulin, ginsenoside Rb3, and genistein showed significant effects in CVDs complications by targeting PPAR-α. With the aim of demonstrating the tremendous chemical variety of natural products targeting PPAR-α in CVDs, this review provides insight into various natural products that can work to prevent CVDs by targeting the PPAR-α receptor along with their detailed mechanism.
Collapse
Affiliation(s)
- Supriya Sharma
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Divya Sharma
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Mahaveer Dhobi
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India.
| | - Dongdong Wang
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON, Canada.
- Department of Medicine, McMaster University, Hamilton, ON, Canada.
| | - Devesh Tewari
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India.
| |
Collapse
|
13
|
Lee IT, Yang CC, Yang CM. Harnessing peroxisome proliferator-activated receptor γ agonists to induce Heme Oxygenase-1: a promising approach for pulmonary inflammatory disorders. Cell Commun Signal 2024; 22:125. [PMID: 38360670 PMCID: PMC10868008 DOI: 10.1186/s12964-024-01501-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/27/2024] [Indexed: 02/17/2024] Open
Abstract
The activation of peroxisome proliferator-activated receptor (PPAR)-γ has been extensively shown to attenuate inflammatory responses in conditions such as asthma, acute lung injury, and acute respiratory distress syndrome, as demonstrated in animal studies. However, the precise molecular mechanisms underlying these inhibitory effects remain largely unknown. The upregulation of heme oxygenase-1 (HO-1) has been shown to confer protective effects, including antioxidant, antiapoptotic, and immunomodulatory effects in vitro and in vivo. PPARγ is highly expressed not only in adipose tissues but also in various other tissues, including the pulmonary system. Thiazolidinediones (TZDs) are highly selective agonists for PPARγ and are used as antihyperglycemic medications. These observations suggest that PPARγ agonists could modulate metabolism and inflammation. Several studies have indicated that PPARγ agonists may serve as potential therapeutic candidates in inflammation-related diseases by upregulating HO-1, which in turn modulates inflammatory responses. In the respiratory system, exposure to external insults triggers the expression of inflammatory molecules, such as cytokines, chemokines, adhesion molecules, matrix metalloproteinases, and reactive oxygen species, leading to the development of pulmonary inflammatory diseases. Previous studies have demonstrated that the upregulation of HO-1 protects tissues and cells from external insults, indicating that the induction of HO-1 by PPARγ agonists could exert protective effects by inhibiting inflammatory signaling pathways and attenuating the development of pulmonary inflammatory diseases. However, the mechanisms underlying TZD-induced HO-1 expression are not well understood. This review aimed to elucidate the molecular mechanisms through which PPARγ agonists induce the expression of HO-1 and explore how they protect against inflammatory and oxidative responses.
Collapse
Affiliation(s)
- I-Ta Lee
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan
| | - Chien-Chung Yang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital at Taoyuan, Taoyuan, 333008, Taiwan
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, 333323, Taiwan
| | - Chuen-Mao Yang
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, 242062, Taiwan.
| |
Collapse
|
14
|
Zhang Y, Xiao B, Liu Y, Wu S, Xiang Q, Xiao Y, Zhao J, Yuan R, Xie K, Li L. Roles of PPAR activation in cancer therapeutic resistance: Implications for combination therapy and drug development. Eur J Pharmacol 2024; 964:176304. [PMID: 38142851 DOI: 10.1016/j.ejphar.2023.176304] [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: 09/17/2023] [Revised: 12/09/2023] [Accepted: 12/21/2023] [Indexed: 12/26/2023]
Abstract
Therapeutic resistance is a major obstacle to successful treatment or effective containment of cancer. Peroxisome proliferator-activated receptors (PPARs) play an essential role in regulating energy homeostasis and determining cell fate. Despite of the pleiotropic roles of PPARs in cancer, numerous studies have suggested their intricate relationship with therapeutic resistance in cancer. In this review, we provided an overview of the roles of excessively activated PPARs in promoting resistance to modern anti-cancer treatments, including chemotherapy, radiotherapy, targeted therapy, and immunotherapy. The mechanisms through which activated PPARs contribute to therapeutic resistance in most cases include metabolic reprogramming, anti-oxidant defense, anti-apoptosis signaling, proliferation-promoting pathways, and induction of an immunosuppressive tumor microenvironment. In addition, we discussed the mechanisms through which activated PPARs lead to multidrug resistance in cancer, including drug efflux, epithelial-to-mesenchymal transition, and acquisition and maintenance of the cancer stem cell phenotype. Preliminary studies investigating the effect of combination therapies with PPAR antagonists have suggested the potential of these antagonists in reversing resistance and facilitating sustained cancer management. These findings will provide a valuable reference for further research on and clinical translation of PPAR-targeting treatment strategies.
Collapse
Affiliation(s)
- Yanxia Zhang
- School of Medicine, The South China University of Technology, Guangzhou, 510006, China; Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Bin Xiao
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Yunduo Liu
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Shunhong Wu
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Qin Xiang
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Yuhan Xiao
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Junxiu Zhao
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Ruanfei Yuan
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Keping Xie
- School of Medicine, The South China University of Technology, Guangzhou, 510006, China.
| | - Linhai Li
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China.
| |
Collapse
|
15
|
Chauhan SS, Casillas AL, Vizzerra AD, Liou H, Clements AN, Flores CE, Prevost CT, Kashatus DF, Snider AJ, Snider JM, Warfel NA. PIM1 drives lipid droplet accumulation to promote proliferation and survival in prostate cancer. Oncogene 2024; 43:406-419. [PMID: 38097734 PMCID: PMC10837079 DOI: 10.1038/s41388-023-02914-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/24/2023] [Accepted: 11/30/2023] [Indexed: 02/04/2024]
Abstract
Lipid droplets (LDs) are dynamic organelles with a neutral lipid core surrounded by a phospholipid monolayer. Solid tumors exhibit LD accumulation, and it is believed that LDs promote cell survival by providing an energy source during energy deprivation. However, the precise mechanisms controlling LD accumulation and utilization in prostate cancer are not well known. Here, we show peroxisome proliferator-activated receptor α (PPARα) acts downstream of PIM1 kinase to accelerate LD accumulation and promote cell proliferation in prostate cancer. Mechanistically, PIM1 inactivates glycogen synthase kinase 3 beta (GSK3β) via serine 9 phosphorylation. GSK3β inhibition stabilizes PPARα and enhances the transcription of genes linked to peroxisomal biogenesis (PEX3 and PEX5) and LD growth (Tip47). The effects of PIM1 on LD accumulation are abrogated with GW6471, a specific inhibitor for PPARα. Notably, LD accumulation downstream of PIM1 provides a significant survival advantage for prostate cancer cells during nutrient stress, such as glucose depletion. Inhibiting PIM reduces LD accumulation in vivo alongside slow tumor growth and proliferation. Furthermore, TKO mice, lacking PIM isoforms, exhibit suppression in circulating triglycerides. Overall, our findings establish PIM1 as an important regulator of LD accumulation through GSK3β-PPARα signaling axis to promote cell proliferation and survival during nutrient stress.
Collapse
Affiliation(s)
- Shailender S Chauhan
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, 85724, USA.
| | - Andrea L Casillas
- Cancer Biology Graduate Program, University of Arizona, Tucson, AZ, 85721, USA
| | - Andres D Vizzerra
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, 85724, USA
| | - Hope Liou
- Cancer Biology Graduate Program, University of Arizona, Tucson, AZ, 85721, USA
| | - Amber N Clements
- Cancer Biology Graduate Program, University of Arizona, Tucson, AZ, 85721, USA
| | - Caitlyn E Flores
- Cancer Biology Graduate Program, University of Arizona, Tucson, AZ, 85721, USA
| | - Christopher T Prevost
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA, 22908, USA
| | - David F Kashatus
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA, 22908, USA
| | - Ashley J Snider
- Department of Nutritional Sciences, College of Agriculture and Life Sciences, University of Arizona, Tucson, AZ, 85721, USA
| | - Justin M Snider
- Department of Nutritional Sciences, College of Agriculture and Life Sciences, University of Arizona, Tucson, AZ, 85721, USA
| | - Noel A Warfel
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, 85724, USA.
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, 85724, USA.
| |
Collapse
|
16
|
Hafiane A. Adiponectin-mediated regulation of the adiponectin cascade in cardiovascular disease: Updates. Biochem Biophys Res Commun 2024; 694:149406. [PMID: 38134479 DOI: 10.1016/j.bbrc.2023.149406] [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: 08/08/2023] [Revised: 12/03/2023] [Accepted: 12/18/2023] [Indexed: 12/24/2023]
Abstract
The endocrine function of white adipose tissue is characterized by the synthesis of one its main hormones: adiponectin. Although the biological role of adiponectin has not been fully defined, clinical and experimental observations have shown that low plasma concentrations of adiponectin participate in the prevalence of insulin resistance and cardiovascular diseases, mainly in obese patients. Adiponectin also exerts its effects on the heart and blood vessels, thereby influencing their physiology. Studying the effects of adiponectin presents some complexities, primarily due to potential cross-interactions and interference with other pathways, such as the AdipoR1/R2 pathways. Under optimal conditions, the activation of the adiponectin cascade may involve signals such as AMPK and PPARα. Interestingly, these pathways may trigger similar responses, such as fatty acid oxidation. Understanding the downstream effectors of these pathways is crucial to comprehend the extent to which adiponectin signaling impacts metabolism. In this review, the aim is to explore the current mechanisms that regulate the adiponectin pathways. Additionally, updates on the major downstream factors involved in adiponectin signaling are provided, specifically in relation to metabolic syndrome and atherosclerosis.
Collapse
Affiliation(s)
- Anouar Hafiane
- Research Institute, McGill University Health Center, Montreal, QC, Canada.
| |
Collapse
|
17
|
Güven B, Sun Q, Wagg CS, Almeida de Oliveira A, Silver H, Persad KL, Onay-Besikci A, Vu J, Oudit GY, Lopaschuk GD. Obesity Is a Major Determinant of Impaired Cardiac Energy Metabolism in Heart Failure with Preserved Ejection Fraction. J Pharmacol Exp Ther 2024; 388:145-155. [PMID: 37977817 DOI: 10.1124/jpet.123.001791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/28/2023] [Accepted: 10/05/2023] [Indexed: 11/19/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a major health problem with limited treatment options. Although optimizing cardiac energy metabolism is a potential approach to treating heart failure, it is poorly understood what alterations in cardiac energy metabolism actually occur in HFpEF. To determine this, we used mice in which HFpEF was induced using an obesity and hypertension HFpEF protocol for 10 weeks. Next, carvedilol, a third-generation β-blocker and a biased agonist that exhibits agonist-like effects through β arrestins by activating extracellular signal-regulated kinase, was used to decrease one of these parameters, namely hypertension. Heart function was evaluated by invasive pressure-volume loops and echocardiography as well as by ex vivo working heart perfusions. Glycolysis and oxidation rates of glucose, fatty acids, and ketones were measured in the isolated working hearts. The development of HFpEF was associated with a dramatic decrease in cardiac glucose oxidation rates, with a parallel increase in palmitate oxidation rates. Carvedilol treatment decreased the development of HFpEF but had no major effect on cardiac energy substrate metabolism. Carvedilol treatment did increase the expression of cardiac β arrestin 2 and proteins involved in mitochondrial biogenesis. Decreasing bodyweight in obese HFpEF mice increased glucose oxidation and improved heart function. This suggests that the dramatic energy metabolic changes in HFpEF mice hearts are primarily due to the obesity component of the HFpEF model. SIGNIFICANCE STATEMENT: Metabolic inflexibility occurs in heart failure with preserved ejection fraction (HFpEF) mice hearts. Lowering blood pressure improves heart function in HFpEF mice with no major effect on energy metabolism. Between hypertension and obesity, the latter appears to have the major role in HFpEF cardiac energetic changes. Carvedilol increases mitochondrial biogenesis and overall energy expenditure in HFpEF hearts.
Collapse
Affiliation(s)
- Berna Güven
- Cardiovascular Research Centre, Department of Pediatrics (B.G., Q.S., C.S.W., H.S., K.L.P., G.D.L.), Department of Medicine, Division of Cardiology (A.A.O., J.V., G.Y.O.), and Mazankowski Alberta Heart Institute (A.A.O., J.V., G.Y.O.), University of Alberta, Edmonton, Canada and Faculty of Pharmacy, Department of Pharmacology, Ankara University, Ankara, Turkey (B.G., A.O.-B.)
| | - Qiuyu Sun
- Cardiovascular Research Centre, Department of Pediatrics (B.G., Q.S., C.S.W., H.S., K.L.P., G.D.L.), Department of Medicine, Division of Cardiology (A.A.O., J.V., G.Y.O.), and Mazankowski Alberta Heart Institute (A.A.O., J.V., G.Y.O.), University of Alberta, Edmonton, Canada and Faculty of Pharmacy, Department of Pharmacology, Ankara University, Ankara, Turkey (B.G., A.O.-B.)
| | - Cory S Wagg
- Cardiovascular Research Centre, Department of Pediatrics (B.G., Q.S., C.S.W., H.S., K.L.P., G.D.L.), Department of Medicine, Division of Cardiology (A.A.O., J.V., G.Y.O.), and Mazankowski Alberta Heart Institute (A.A.O., J.V., G.Y.O.), University of Alberta, Edmonton, Canada and Faculty of Pharmacy, Department of Pharmacology, Ankara University, Ankara, Turkey (B.G., A.O.-B.)
| | - Amanda Almeida de Oliveira
- Cardiovascular Research Centre, Department of Pediatrics (B.G., Q.S., C.S.W., H.S., K.L.P., G.D.L.), Department of Medicine, Division of Cardiology (A.A.O., J.V., G.Y.O.), and Mazankowski Alberta Heart Institute (A.A.O., J.V., G.Y.O.), University of Alberta, Edmonton, Canada and Faculty of Pharmacy, Department of Pharmacology, Ankara University, Ankara, Turkey (B.G., A.O.-B.)
| | - Heidi Silver
- Cardiovascular Research Centre, Department of Pediatrics (B.G., Q.S., C.S.W., H.S., K.L.P., G.D.L.), Department of Medicine, Division of Cardiology (A.A.O., J.V., G.Y.O.), and Mazankowski Alberta Heart Institute (A.A.O., J.V., G.Y.O.), University of Alberta, Edmonton, Canada and Faculty of Pharmacy, Department of Pharmacology, Ankara University, Ankara, Turkey (B.G., A.O.-B.)
| | - Kaya L Persad
- Cardiovascular Research Centre, Department of Pediatrics (B.G., Q.S., C.S.W., H.S., K.L.P., G.D.L.), Department of Medicine, Division of Cardiology (A.A.O., J.V., G.Y.O.), and Mazankowski Alberta Heart Institute (A.A.O., J.V., G.Y.O.), University of Alberta, Edmonton, Canada and Faculty of Pharmacy, Department of Pharmacology, Ankara University, Ankara, Turkey (B.G., A.O.-B.)
| | - Arzu Onay-Besikci
- Cardiovascular Research Centre, Department of Pediatrics (B.G., Q.S., C.S.W., H.S., K.L.P., G.D.L.), Department of Medicine, Division of Cardiology (A.A.O., J.V., G.Y.O.), and Mazankowski Alberta Heart Institute (A.A.O., J.V., G.Y.O.), University of Alberta, Edmonton, Canada and Faculty of Pharmacy, Department of Pharmacology, Ankara University, Ankara, Turkey (B.G., A.O.-B.)
| | - Jennie Vu
- Cardiovascular Research Centre, Department of Pediatrics (B.G., Q.S., C.S.W., H.S., K.L.P., G.D.L.), Department of Medicine, Division of Cardiology (A.A.O., J.V., G.Y.O.), and Mazankowski Alberta Heart Institute (A.A.O., J.V., G.Y.O.), University of Alberta, Edmonton, Canada and Faculty of Pharmacy, Department of Pharmacology, Ankara University, Ankara, Turkey (B.G., A.O.-B.)
| | - Gavin Y Oudit
- Cardiovascular Research Centre, Department of Pediatrics (B.G., Q.S., C.S.W., H.S., K.L.P., G.D.L.), Department of Medicine, Division of Cardiology (A.A.O., J.V., G.Y.O.), and Mazankowski Alberta Heart Institute (A.A.O., J.V., G.Y.O.), University of Alberta, Edmonton, Canada and Faculty of Pharmacy, Department of Pharmacology, Ankara University, Ankara, Turkey (B.G., A.O.-B.)
| | - Gary D Lopaschuk
- Cardiovascular Research Centre, Department of Pediatrics (B.G., Q.S., C.S.W., H.S., K.L.P., G.D.L.), Department of Medicine, Division of Cardiology (A.A.O., J.V., G.Y.O.), and Mazankowski Alberta Heart Institute (A.A.O., J.V., G.Y.O.), University of Alberta, Edmonton, Canada and Faculty of Pharmacy, Department of Pharmacology, Ankara University, Ankara, Turkey (B.G., A.O.-B.)
| |
Collapse
|
18
|
Florke Gee RR, Huber AD, Wu J, Bajpai R, Loughran AJ, Pruett-Miller SM, Chen T. The F-box-only protein 44 regulates pregnane X receptor protein level by ubiquitination and degradation. Acta Pharm Sin B 2023; 13:4523-4534. [PMID: 37969738 PMCID: PMC10638512 DOI: 10.1016/j.apsb.2023.07.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/24/2023] [Accepted: 06/13/2023] [Indexed: 11/17/2023] Open
Abstract
Pregnane X receptor (PXR) is a ligand-activated nuclear receptor that transcriptionally upregulates drug-metabolizing enzymes [e.g., cytochrome P450 3A4 (CYP3A4)] and transporters. Although the regulation of PXR target genes is well-characterized, less is known about the regulation of PXR protein level. By screening an RNAi library, we identified the F-box-only protein 44 (FBXO44) as a novel E3 ligase for PXR. PXR abundance increases upon knockdown of FBXO44, and, inversely, decreases upon overexpression of FBXO44. Further analysis revealed that FBXO44 interacts with PXR, leading to its ubiquitination and proteasomal degradation, and we determined that the F-box associated domain of FBXO44 and the ligand binding domain of PXR are required for the functional interaction. In summary, FBXO44 regulates PXR protein abundance, which has downstream consequences for CYP3A4 levels and drug-drug interactions. The results of this study provide new insight into the molecular mechanisms that regulate PXR protein level and activity and suggest the importance of considering how modulating E3 ubiquitin ligase activities will affect PXR-mediated drug metabolism.
Collapse
Affiliation(s)
- Rebecca R. Florke Gee
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Andrew D. Huber
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jing Wu
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Richa Bajpai
- Center for Advanced Genome Engineering and Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Allister J. Loughran
- Center for Advanced Genome Engineering and Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Shondra M. Pruett-Miller
- Center for Advanced Genome Engineering and Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
19
|
Boubertakh B, Courtemanche O, Marsolais D, Di Marzo V, Silvestri C. New role for the anandamide metabolite prostaglandin F 2α ethanolamide: Rolling preadipocyte proliferation. J Lipid Res 2023; 64:100444. [PMID: 37730163 PMCID: PMC10622703 DOI: 10.1016/j.jlr.2023.100444] [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: 05/13/2023] [Revised: 08/24/2023] [Accepted: 09/11/2023] [Indexed: 09/22/2023] Open
Abstract
White adipose tissue regulation is key to metabolic health, yet still perplexing. The chief endocannabinoid anandamide metabolite, prostaglandin F2α ethanolamide (PGF2αEA), inhibits adipogenesis, that is, the formation of mature adipocytes. We observed that adipocyte progenitor cells-preadipocytes-following treatment with PGF2αEA yielded larger pellet sizes. Thus, we hypothesized that PGF2αEA might augment preadipocyte proliferation. Cell viability MTT and crystal violet assays, cell counting, and 5-bromo-2'-deoxyuridine incorporation in cell proliferation ELISA analyses confirmed our prediction. Additionally, we discovered that PGF2αEA promotes cell cycle progression through suppression of the expression of cell cycle inhibitors, p21 and p27, as shown by flow cytometry and qPCR. Enticingly, concentrations of this compound that showed no visible effect on cell proliferation or basal transcriptional activity of peroxisome proliferator-activated receptor gamma could, in contrast, reverse the anti-proliferative and peroxisome proliferator-activated receptor gamma-transcription activating effects of rosiglitazone (Rosi). MTT and luciferase reporter examinations supported this finding. The PGF2αEA pharmaceutical analog, bimatoprost, was also investigated and showed very similar effects. Importantly, we suggest the implication of the mitogen-activated protein kinase pathway in these effects, as they were blocked by the selective mitogen-activated protein kinase kinase inhibitor, PD98059. We propose that PGF2αEA is a pivotal regulator of white adipose tissue plasticity, acting as a regulator of the preadipocyte pool in adipose tissue.
Collapse
Affiliation(s)
- Besma Boubertakh
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Département de médecine, Faculté de Médecine, Université Laval, Québec, Canada; Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Centre NUTRISS, Université Laval, Québec, Canada; Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Québec, Canada
| | - Olivier Courtemanche
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Département de médecine, Faculté de Médecine, Université Laval, Québec, Canada
| | - David Marsolais
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Département de médecine, Faculté de Médecine, Université Laval, Québec, Canada
| | - Vincenzo Di Marzo
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Département de médecine, Faculté de Médecine, Université Laval, Québec, Canada; Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Centre NUTRISS, Université Laval, Québec, Canada; Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Québec, Canada; École de Nutrition, Faculté des Sciences de l'Agriculture et de l'Alimentation (FSAA), Université Laval, Québec, Canada
| | - Cristoforo Silvestri
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Département de médecine, Faculté de Médecine, Université Laval, Québec, Canada; Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Centre NUTRISS, Université Laval, Québec, Canada; Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Québec, Canada.
| |
Collapse
|
20
|
Fuior EV, Zvintzou E, Filippatos T, Giannatou K, Mparnia V, Simionescu M, Gafencu AV, Kypreos KE. Peroxisome Proliferator-Activated Receptor α in Lipoprotein Metabolism and Atherosclerotic Cardiovascular Disease. Biomedicines 2023; 11:2696. [PMID: 37893070 PMCID: PMC10604751 DOI: 10.3390/biomedicines11102696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/26/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are a group of ligand-binding transcription factors with pivotal action in regulating pleiotropic signaling pathways of energetic metabolism, immune responses and cell proliferation and differentiation. A significant body of evidence indicates that the PPARα receptor is an important modulator of plasma lipid and lipoprotein metabolism, with pluripotent effects influencing the lipid and apolipoprotein cargo of both atherogenic and antiatherogenic lipoproteins and their functionality. Clinical evidence supports an important role of PPARα agonists (fibric acid derivatives) in the treatment of hypertriglyceridemia and/or low high-density lipoprotein (HDL) cholesterol levels, although the effects of clinical trials are contradictory and point to a reduction in the risk of nonfatal and fatal myocardial infarction events. In this manuscript, we provide an up-to-date critical review of the existing relevant literature.
Collapse
Affiliation(s)
- Elena Valeria Fuior
- Institute of Cellular Biology and Pathology, “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (E.V.F.); (E.Z.); (M.S.)
| | - Evangelia Zvintzou
- Institute of Cellular Biology and Pathology, “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (E.V.F.); (E.Z.); (M.S.)
- Pharmacology Laboratory, Department of Medicine, University of Patras, 26500 Rio Achaias, Greece; (K.G.); (V.M.)
| | - Theodosios Filippatos
- Internal Medicine Clinic, Department of Medicine, University of Crete, 71500 Heraklion, Greece;
| | - Katerina Giannatou
- Pharmacology Laboratory, Department of Medicine, University of Patras, 26500 Rio Achaias, Greece; (K.G.); (V.M.)
| | - Victoria Mparnia
- Pharmacology Laboratory, Department of Medicine, University of Patras, 26500 Rio Achaias, Greece; (K.G.); (V.M.)
| | - Maya Simionescu
- Institute of Cellular Biology and Pathology, “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (E.V.F.); (E.Z.); (M.S.)
| | - Anca Violeta Gafencu
- Institute of Cellular Biology and Pathology, “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (E.V.F.); (E.Z.); (M.S.)
| | - Kyriakos E. Kypreos
- Institute of Cellular Biology and Pathology, “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (E.V.F.); (E.Z.); (M.S.)
- Pharmacology Laboratory, Department of Medicine, University of Patras, 26500 Rio Achaias, Greece; (K.G.); (V.M.)
- Department of Life Sciences, School of Sciences, European University Cyprus, 2404 Nicosia, Cyprus
| |
Collapse
|
21
|
Ballav S, Ranjan A, Basu S. Partial Activation of PPAR-γ by Synthesized Quercetin Derivatives Modulates TGF-β1-Induced EMT in Lung Cancer Cells. Adv Biol (Weinh) 2023; 7:e2300037. [PMID: 37042092 DOI: 10.1002/adbi.202300037] [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: 01/23/2023] [Revised: 03/10/2023] [Indexed: 04/13/2023]
Abstract
Non-small cell lung cancer (NSCLC) has a very low survival rate due to poor response to chemotherapy and late detection. Epithelial to mesenchymal transition (EMT) is regarded as a major contributor to drive metastasis during NSCLC progression. Towards this, transforming growth factor-beta 1 (TGF-β1) is the key driver that endows cancer cells with increased aggressiveness. Recently, this group synthesized a series of Schiff base quercetin derivatives (QDs) and ascertained their effectiveness on EMT markers of A549 cell line. This study evidenced that the EMT process is counteracted via the partial activation of a nuclear hormone receptor, Peroxisome proliferator-activated receptor (PPAR)-γ through QDs. Here, that work is extended to investigate the interplay between PPAR-γ partial activation and TGF-β1-induced EMT in human lung cancer A549 cells. The results reveal that TGF-β1 plays a critical role in suppressing PPAR-γ, which is markedly reversed and increased by partial agonists: QUE2FH and QUESH at both protein and transcriptional levels. The partial agonists not only stimulate PPAR-γ in a balanced manner but also prevent the loss of E-cadherin and acquisition of TGF-β1-induced mesenchymal markers (Snail, Slug, Vimentin, and Zeb-1). Subsequently, the effects are accompanied by attenuation of TGF-β1-induced migratory ability of A549 cells.
Collapse
Affiliation(s)
- Sangeeta Ballav
- Cancer and Translational Research Centre, Dr. D.Y. Patil Biotechnology and Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Tathawade, Pune, Maharashtra, 411 033, India
| | - Amit Ranjan
- Cancer and Translational Research Centre, Dr. D.Y. Patil Biotechnology and Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Tathawade, Pune, Maharashtra, 411 033, India
| | - Soumya Basu
- Cancer and Translational Research Centre, Dr. D.Y. Patil Biotechnology and Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Tathawade, Pune, Maharashtra, 411 033, India
| |
Collapse
|
22
|
Kumar S, Chhimwal J, Kumar S, Singh R, Patial V, Purohit R, Padwad YS. Phloretin and phlorizin mitigates inflammatory stress and alleviate adipose and hepatic insulin resistance by abrogating PPARγ S273-Cdk5 interaction in type 2 diabetic mice. Life Sci 2023; 322:121668. [PMID: 37023949 DOI: 10.1016/j.lfs.2023.121668] [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: 03/01/2023] [Revised: 03/31/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023]
Abstract
AIMS The rising prevalence of type 2 diabetes mellitus (T2DM) and accompanying insulin resistance is alarming globally. Natural and synthetic agonists of PPARγ are potentially attractive candidates for diabetics and are known to efficiently reverse adipose and hepatic insulin resistance, but related side effects and escalating costs are the causes of concern. Therefore, targeting PPARγ with natural ligands is advantageous and promising approach for the better management of T2DM. The present research aimed to assess the antidiabetic potential of phenolics Phloretin (PTN) and Phlorizin (PZN) in type 2 diabetic mice. MAIN METHODS In silico docking was performed to check the effect of PTN and PZN on PPARγ S273-Cdk5 interactions. The docking results were further validated in preclinical settings by utilizing a mice model of high fat diet-induced T2DM. KEY FINDINGS Computational docking and further MD-simulation data revealed that PTN and PZN inhibited the activation of Cdk5, thereby blocking the phosphorylation of PPARγ. Our in vivo results further demonstrated that PTN and PZN administration significantly improved the secretory functions of adipocytes by increasing adiponectin and reducing inflammatory cytokine levels, which ultimately reduced the hyperglycaemic index. Additionally, combined treatment of PTN and PZN decreased in vivo adipocyte expansion and increased Glut4 expression in adipose tissues. Furthermore, PTN and PZN treatment reduced hepatic insulin resistance by modulating lipid metabolism and inflammatory markers. SIGNIFICANCE In summary, our findings strongly imply that PTN and PZN are candidates as nutraceuticals in the management of comorbidities related to diabetes and its complications.
Collapse
Affiliation(s)
- Shiv Kumar
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, U.P., India
| | - Jyoti Chhimwal
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, U.P., India
| | - Suresh Kumar
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, U.P., India
| | - Rahul Singh
- Structural Bioinformatics Lab, Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, U.P., India
| | - Vikram Patial
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, U.P., India
| | - Rituraj Purohit
- Structural Bioinformatics Lab, Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, U.P., India.
| | - Yogendra S Padwad
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, U.P., India.
| |
Collapse
|
23
|
Chen Q, Deng X, Zhang K, Kang Y, Jiao M, Zhang J, Wang C, Li F. Changes to PUFA-PPAR pathway during mesaconitine induced myocardial coagulative necrosis. Food Chem Toxicol 2023; 177:113831. [PMID: 37182599 DOI: 10.1016/j.fct.2023.113831] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 05/16/2023]
Abstract
Coagulation necrosis is characterized by the denaturation of structural proteins and lysosomal enzymes; its occurrence in myocardium can lead to heart failure. Current studies on myocardial injury primarily focus on inflammation, hypertrophy, and hemorrhage, while those on myocardial coagulation necrosis are still limited. Mesaconitine (MA), a C19 diester diterpenoid alkaloid derived from Aconitum carmichaelii Debx, has strong cardiotoxicity. During this study, the myocardial cells of SD rats showed significant coagulative necrosis after 6 days of oral administration of MA at a dose of 1.2 mg/kg/day. Investigations of its biological mechanism showed abnormal levels of polyunsaturated fatty acids (PUFAs) and Peroxisome proliferator activated receptors Alpha (PPARα) pathway related protein. Moreover, MA affected the PPARα signaling pathway through interactions with proteins such as POR, TFAM and GPD1, indirectly indicating that these above proteins are important targets for blocking myocardial coagulative necrosis. This study thus discusses the effects of the use of cardiotoxic compound, MA, to initiate myocardial coagulative necrosis and its associated toxic mechanisms.
Collapse
Affiliation(s)
- Qian Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing, 102488, China.
| | - Xinqi Deng
- School of Life Sciences, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing, 102488, China.
| | - Kai Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing, 102488, China.
| | - Yingquan Kang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing, 102488, China.
| | - Mingjie Jiao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing, 102488, China.
| | - Jia Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing, 102488, China.
| | - Chunguo Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing, 102488, China.
| | - Fei Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing, 102488, China.
| |
Collapse
|
24
|
Cizkova K, Tauber Z. Fibrates Affect Levels of Phosphorylated p38 in Intestinal Cells in a Differentiation-Dependent Manner. Int J Mol Sci 2023; 24:ijms24097695. [PMID: 37175404 PMCID: PMC10178720 DOI: 10.3390/ijms24097695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
Fibrates are widely used hypolipidaemic agents that act as ligands of the peroxisome proliferator-activated receptor α (PPARα). p38 is a protein kinase that is mainly activated by environmental and genotoxic stress. We investigated the effect of the PPARα activators fenofibrate and WY-14643 and the PPARα inhibitor GW6471 on the levels of activated p38 (p-p38) in the colorectal cancer cell lines HT-29 and Caco2 in relation to their differentiation status. Fibrates increased p-p38 in undifferentiated HT-29 cells, whereas in other cases p-p38 expression was decreased. HT-29 cells showed p-p38 predominantly in the cytoplasm, whereas Caco2 cells showed higher nuclear positivity. The effect of fibrates may depend on the differentiation status of the cell, as differentiated HT-29 and undifferentiated Caco2 cells share similar characteristics in terms of villin, CYP2J2, and soluble epoxide hydrolase (sEH) expression. In human colorectal carcinoma, higher levels of p-p38 were detected in the cytoplasm, whereas in normal colonic surface epithelium, p-p38 showed nuclear positivity. The decrease in p-p38 positivity was associated with a decrease in sEH, consistent with in vitro results. In conclusion, fibrates affect the level of p-p38, but its exact role in the process of carcinogenesis remains unclear and further research is needed in this area.
Collapse
Affiliation(s)
- Katerina Cizkova
- Department of Histology and Embryology, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic
| | - Zdenek Tauber
- Department of Histology and Embryology, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic
| |
Collapse
|
25
|
Zhang K, Yang X, Zheng M, Ning Y, Zhang S. Acetylated-PPARγ expression is regulated by different P53 genotypes associated with the adipogenic differentiation of polyploid giant cancer cells with daughter cells. Cancer Biol Med 2023; 20:56-76. [PMID: 36647790 PMCID: PMC9843444 DOI: 10.20892/j.issn.2095-3941.2022.0432] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/04/2022] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVE Polyploid giant cancer cells (PGCCs) with daughter cells express epithelial-mesenchymal transition (EMT)-associated proteins. Highly malignant tumor cells with EMT properties can transdifferentiate into mature tumor cells. In this study, we elucidated the potential for, and underlying mechanism of, adipogenic differentiation of PGCCs with daughter cells (PDCs). METHODS Cobalt chloride was used to induce PGCC formation in HEY (wild-type P53) and MDA-MB-231 (mutant P53) cells; these cells were then cultured in adipogenic differentiation medium. Oil red O staining was used to confirm adipogenic differentiation, and the cell cycle was detected with flow cytometry. The expression of adipogenic differentiation-associated proteins and P300 histone acetyltransferase activity were compared before and after adipogenic differentiation. Animal xenograft models were used to confirm the adipogenic differentiation of PDCs. RESULTS PDCs transdifferentiated into functional adipocytes. Two different cell cycle distributions were observed in PDCs after adipogenic differentiation. The expression levels of PPARγ, Ace-PPARγ, and Ace-P53 were higher in PDCs after adipogenic differentiation than in cells before adipogenic differentiation. Ace-PPARγ and FABP4 expression increased in HEY cells and decreased in MDA-MB-231 PDCs after p53 knockdown. A485 treatment increased Ace-P53, Ace-PPARγ, and FABP4 expression in HEY PDCs by inhibiting SUMOylation of P53. In MDA-MB-231 PDCs, A485 treatment decreased Ace-P53, Ace-PPARγ, and FABP4 expression. Animal experiments also confirmed the adipogenic differentiation of PDCs. CONCLUSIONS Acetylation of P53 and PPARγ plays an important role in the adipogenic differentiation of PDCs.
Collapse
Affiliation(s)
- Kexin Zhang
- Tianjin Union Medical Center, Tianjin 300122, China
| | - Xiaohui Yang
- Nankai University School of Medicine, Nankai University, Tianjin 300071, China
| | | | - Yidi Ning
- Nankai University School of Medicine, Nankai University, Tianjin 300071, China
| | - Shiwu Zhang
- Tianjin Union Medical Center, Tianjin 300122, China
| |
Collapse
|
26
|
Roth D, Benz J, Grether U, Dietz M. Homogeneous Time-Resolved Fluorescence Resonance Energy Transfer (TR-FRET) Cofactor Recruitment Assay for PPARα and PPARγ. Methods Mol Biol 2023; 2576:155-169. [PMID: 36152184 DOI: 10.1007/978-1-0716-2728-0_12] [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] [Indexed: 06/16/2023]
Abstract
Peroxisome proliferator-activated receptors are a family of nuclear hormone receptors that control the expression of genes involved in a variety of physiologic processes, through heterodimerization with retinoid X receptor and complex formation with various cofactors. The specific cofactors recruited to PPAR-RXR complexes in response to different ligands lead to major differences in the transactivation of target genes. We developed a cofactor recruitment assay that is based on an europium-labeled anti-GST antibody and streptavidin-APC leading to a fluorescence resonance energy transfer signal. This assay allows for the determination of unique agonistic profiles in terms of potency and co-activator motif. Hence, it is a valuable drug discovery tool to support hit finding and lead optimization campaigns, enabling the characterization of next generation PPAR agonists.
Collapse
Affiliation(s)
- Doris Roth
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland.
| | - Jörg Benz
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Uwe Grether
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Michel Dietz
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| |
Collapse
|
27
|
The Role of PPARs in Breast Cancer. Cells 2022; 12:cells12010130. [PMID: 36611922 PMCID: PMC9818187 DOI: 10.3390/cells12010130] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/07/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022] Open
Abstract
Breast cancer is a malignant tumor with high morbidity and lethality. Its pathogenesis is related to the abnormal expression of many genes. The peroxisome proliferator-activated receptors (PPARs) are a class of ligand-dependent transcription factors in the nuclear receptor superfamily. They can regulate the transcription of a large number of target genes, which are involved in life activities such as cell proliferation, differentiation, metabolism, and apoptosis, and regulate physiological processes such as glucose metabolism, lipid metabolism, inflammation, and wound healing. Further, the changes in its expression are associated with various diseases, including breast cancer. The experimental reports related to "PPAR" and "breast cancer" were retrieved from PubMed since the discovery of PPARs and summarized in this paper. This review (1) analyzed the roles and potential molecular mechanisms of non-coordinated and ligand-activated subtypes of PPARs in breast cancer progression; (2) discussed the correlations between PPARs and estrogen receptors (ERs) as the nuclear receptor superfamily; and (3) investigated the interaction between PPARs and key regulators in several signaling pathways. As a result, this paper identifies PPARs as targets for breast cancer prevention and treatment in order to provide more evidence for the synthesis of new drugs targeting PPARs or the search for new drug combination treatments.
Collapse
|
28
|
Benatzy Y, Palmer MA, Brüne B. Arachidonate 15-lipoxygenase type B: Regulation, function, and its role in pathophysiology. Front Pharmacol 2022; 13:1042420. [PMID: 36438817 PMCID: PMC9682198 DOI: 10.3389/fphar.2022.1042420] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/26/2022] [Indexed: 10/30/2023] Open
Abstract
As a lipoxygenase (LOX), arachidonate 15-lipoxygenase type B (ALOX15B) peroxidizes polyenoic fatty acids (PUFAs) including arachidonic acid (AA), eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), and linoleic acid (LA) to their corresponding fatty acid hydroperoxides. Distinctive to ALOX15B, fatty acid oxygenation occurs with positional specificity, catalyzed by the non-heme iron containing active site, and in addition to free PUFAs, membrane-esterified fatty acids serve as substrates for ALOX15B. Like other LOX enzymes, ALOX15B is linked to the formation of specialized pro-resolving lipid mediators (SPMs), and altered expression is apparent in various inflammatory diseases such as asthma, psoriasis, and atherosclerosis. In primary human macrophages, ALOX15B expression is associated with cellular cholesterol homeostasis and is induced by hypoxia. Like in inflammation, the role of ALOX15B in cancer is inconclusive. In prostate and breast carcinomas, ALOX15B is attributed a tumor-suppressive role, whereas in colorectal cancer, ALOX15B expression is associated with a poorer prognosis. As the biological function of ALOX15B remains an open question, this review aims to provide a comprehensive overview of the current state of research related to ALOX15B.
Collapse
Affiliation(s)
- Yvonne Benatzy
- Faculty of Medicine, Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany
| | - Megan A. Palmer
- Faculty of Medicine, Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany
| | - Bernhard Brüne
- Faculty of Medicine, Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt, Germany
| |
Collapse
|
29
|
The Potential Role of PPARs in the Fetal Origins of Adult Disease. Cells 2022; 11:cells11213474. [PMID: 36359869 PMCID: PMC9653757 DOI: 10.3390/cells11213474] [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: 09/25/2022] [Revised: 10/19/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
Abstract
The fetal origins of adult disease (FOAD) hypothesis holds that events during early development have a profound impact on one’s risk for the development of future adult disease. Studies from humans and animals have demonstrated that many diseases can begin in childhood and are caused by a variety of early life traumas, including maternal malnutrition, maternal disease conditions, lifestyle changes, exposure to toxins/chemicals, improper medication during pregnancy, and so on. Recently, the roles of Peroxisome proliferator-activated receptors (PPARs) in FOAD have been increasingly appreciated due to their wide variety of biological actions. PPARs are members of the nuclear hormone receptor subfamily, consisting of three distinct subtypes: PPARα, β/δ, and γ, highly expressed in the reproductive tissues. By controlling the maturation of the oocyte, ovulation, implantation of the embryo, development of the placenta, and male fertility, the PPARs play a crucial role in the transition from embryo to fetus in developing mammals. Exposure to adverse events in early life exerts a profound influence on the methylation pattern of PPARs in offspring organs, which can affect development and health throughout the life course, and even across generations. In this review, we summarize the latest research on PPARs in the area of FOAD, highlight the important role of PPARs in FOAD, and provide a potential strategy for early prevention of FOAD.
Collapse
|
30
|
Wetten A, Jones DEJ, Dyson JK. Seladelpar: an investigational drug for the treatment of early-stage primary biliary cholangitis (PBC). Expert Opin Investig Drugs 2022; 31:1101-1107. [PMID: 36194037 DOI: 10.1080/13543784.2022.2130750] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Generic fibrates are used off-label as add-in therapy for the management of primary biliary cholangitis (PBC) but with unproven long-term liver-related survival benefits. The recently developed fibrate, seladelpar, has shown promising results in clinical trials, but these outcomes have been previously marred by safety concerns. AREAS COVERED We summarize existing treatment options in PBC and evaluate current trial data for seladelpar in relation to liver biochemistry, symptomology, and safety. EXPERT OPINION Seladelpar leads to marked improvement in liver biochemistry and may improve symptoms. Safety concerns around liver toxicity appear to have been addressed. With likely increasing evidence compared to existing off-label fibrates, seladelpar has the potential as an attractive future second-line agent in PBC.
Collapse
Affiliation(s)
- Aaron Wetten
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, UK.,Liver unit, Freeman Hospital, Newcastle-upon-Tyne, UK
| | - David Emrys Jeffreys Jones
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, UK.,Liver unit, Freeman Hospital, Newcastle-upon-Tyne, UK
| | - Jessica Katharine Dyson
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, UK.,Liver unit, Freeman Hospital, Newcastle-upon-Tyne, UK
| |
Collapse
|
31
|
Mori C, Lee JY, Tokumoto M, Satoh M. Cadmium Toxicity Is Regulated by Peroxisome Proliferator-Activated Receptor δ in Human Proximal Tubular Cells. Int J Mol Sci 2022; 23:ijms23158652. [PMID: 35955783 PMCID: PMC9369238 DOI: 10.3390/ijms23158652] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/26/2022] [Accepted: 08/02/2022] [Indexed: 12/13/2022] Open
Abstract
Cadmium (Cd) is a toxic heavy metal that is widely present in the environment. Renal proximal tubule disorder is the main symptom of Cd chronic poisoning. Our previous study demonstrated that Cd inhibits the total activities of peroxisome proliferator-activated receptor (PPAR) transcription factors in human and rat proximal tubular cells. In this study, we investigated the involvement of PPAR in Cd renal toxicity using the HK-2 human proximal tubular cell line. Among PPAR isoform genes, only PPARD knockdown significantly showed resistance to Cd toxicity in HK-2 cells. The transcriptional activity of PPARδ was decreased not only by PPARD knockdown but also by Cd treatment. DNA microarray analysis showed that PPARD knockdown changed the expression of apoptosis-related genes in HK-2 cells. PPARD knockdown decreased apoptosis signals and caspase-3 activity induced by Cd treatment. PPARD knockdown did not affect the intracellular Cd level after Cd treatment. These results suggest that PPARδ plays a critical role in the modification of susceptibility to Cd renal toxicity and that the apoptosis pathway may be involved in PPARδ-related Cd toxicity.
Collapse
|
32
|
Cercillieux A, Ciarlo E, Canto C. Balancing NAD + deficits with nicotinamide riboside: therapeutic possibilities and limitations. Cell Mol Life Sci 2022; 79:463. [PMID: 35918544 PMCID: PMC9345839 DOI: 10.1007/s00018-022-04499-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/20/2022] [Accepted: 07/20/2022] [Indexed: 12/21/2022]
Abstract
Alterations in cellular nicotinamide adenine dinucleotide (NAD+) levels have been observed in multiple lifestyle and age-related medical conditions. This has led to the hypothesis that dietary supplementation with NAD+ precursors, or vitamin B3s, could exert health benefits. Among the different molecules that can act as NAD+ precursors, Nicotinamide Riboside (NR) has gained most attention due to its success in alleviating and treating disease conditions at the pre-clinical level. However, the clinical outcomes for NR supplementation strategies have not yet met the expectations generated in mouse models. In this review we aim to provide a comprehensive view on NAD+ biology, what causes NAD+ deficits and the journey of NR from its discovery to its clinical development. We also discuss what are the current limitations in NR-based therapies and potential ways to overcome them. Overall, this review will not only provide tools to understand NAD+ biology and assess its changes in disease situations, but also to decide which NAD+ precursor could have the best therapeutic potential.
Collapse
Affiliation(s)
- Angelique Cercillieux
- Nestlé Institute of Health Sciences, Nestlé Research Ltd., EPFL Campus, Innovation Park, Building G, 1015, Lausanne, Switzerland
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Eleonora Ciarlo
- Nestlé Institute of Health Sciences, Nestlé Research Ltd., EPFL Campus, Innovation Park, Building G, 1015, Lausanne, Switzerland
| | - Carles Canto
- Nestlé Institute of Health Sciences, Nestlé Research Ltd., EPFL Campus, Innovation Park, Building G, 1015, Lausanne, Switzerland.
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland.
| |
Collapse
|
33
|
Dong S, Li S, Wang X, Liang S, Zhang W, Li L, Xu Q, Shi B, Cheng Z, Zhang X, Zhong M, Zhang G, Hu S. CD147 Mediates 5-Fluorouracil Resistance in Colorectal Cancer by Reprogramming Glycolipid Metabolism. Front Oncol 2022; 12:813852. [PMID: 35898887 PMCID: PMC9309564 DOI: 10.3389/fonc.2022.813852] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Chemoresistance against 5-fluorouracil (5-FU) is a major issue for colorectal cancer (CRC) patients. Increasing evidence for the roles of CD147 in glycolipid metabolic reprogramming and chemoresistance of tumor cells has emerged in recent years. However, whether CD147 contributes to 5-FU resistance in CRC and the role of abnormal glycolipid metabolism in this process remain poorly understood. We analyzed CD147 expression in primary tumor samples of CRC patients and found that upregulated CD147 correlated with decreased 5-FU chemosensitivity and an unfavorable prognosis of CRC patients. Moreover, in vivo and in vitro experiments confirmed that CD147 regulates glycolipid metabolism through two separate pathways. Mechanistically, CD147 upregulates HIF-1α-mediated glycolysis by activating the PI3K/AKT/mTOR pathway and CD147 also attenuates PPARα-mediated fatty acid oxidation by activation of the MAPK pathway. Most importantly, we found that CD147 confers 5-FU resistance in CRC via these glycolipid metabolic signatures. Our results demonstrated that CD147 is a potential 5-FU resistance biomarker for CRC patients and a candidate therapeutic target to restore 5-FU sensitivity of 5-FU-resistant CRC by remodeling glycolipid metabolism.
Collapse
Affiliation(s)
- Shuohui Dong
- Department of General Surgery, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Songhan Li
- Department of General Surgery, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaoyan Wang
- Department of Neonatology, Weifang Yidu Central Hospital, Weifang, China
| | - Shuo Liang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial Ear-nose-throat (ENT) Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wenjie Zhang
- Department of General Surgery, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Linchuan Li
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Qian Xu
- Department of General Surgery, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bowen Shi
- Department of General Surgery, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhiqiang Cheng
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiang Zhang
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Mingwei Zhong
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Guangyong Zhang
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Sanyuan Hu
- Department of General Surgery, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: SanYuan Hu,
| |
Collapse
|
34
|
Guixé‐Muntet S, Biquard L, Szabo G, Dufour J, Tacke F, Francque S, Rautou P, Gracia‐Sancho J. Review article: vascular effects of PPARs in the context of NASH. Aliment Pharmacol Ther 2022; 56:209-223. [PMID: 35661191 PMCID: PMC9328268 DOI: 10.1111/apt.17046] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/04/2021] [Accepted: 05/08/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors known to regulate glucose and fatty acid metabolism, inflammation, endothelial function and fibrosis. PPAR isoforms have been extensively studied in metabolic diseases, including type 2 diabetes and cardiovascular diseases. Recent data extend the key role of PPARs to liver diseases coursing with vascular dysfunction, including nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH). AIM This review summarises and discusses the pathobiological role of PPARs in cardiovascular diseases with a special focus on their impact and therapeutic potential in NAFLD and NASH. RESULTS AND CONCLUSIONS PPARs may be attractive for the treatment of NASH due to their liver-specific effects but also because of their efficacy in improving cardiovascular outcomes, which may later impact liver disease. Assessment of cardiovascular disease in the context of NASH trials is, therefore, of the utmost importance, both from a safety and efficacy perspective.
Collapse
Affiliation(s)
- Sergi Guixé‐Muntet
- Liver Vascular Biology Research GroupIDIBAPS Biomedical Research Institute & CIBEREHDBarcelonaSpain
| | - Louise Biquard
- Université de Paris, Inserm, CNRSCentre de recherche sur l'InflammationUMR1149ParisFrance
| | - Gyongyi Szabo
- Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Jean‐François Dufour
- Department of Visceral Surgery and Medicine & Department for Biomedical ResearchInselspital, University of BernBernSwitzerland
| | - Frank Tacke
- Department of Hepatology & GastroenterologyCharité Universitätsmedizin Berlin, Campus Virchow‐Klinikum (CVK) and Campus Charité Mitte (CCM)BerlinGermany
| | - Sven Francque
- Department of Gastroenterology and HepatologyAntwerp University HospitalAntwerpBelgium,Translational Sciences in Inflammation and ImmunologyInflaMed Centre of Excellence, Laboratory of Experimental Medicine and Paediatrics, Faculty of Medicine and Health Sciences, University of AntwerpAntwerpBelgium
| | - Pierre‐Emmanuel Rautou
- Université de Paris, AP‐HP, Hôpital Beaujon, Service d'Hépatologie, DMU DIGESTCentre de Référence des Maladies Vasculaires du Foie, FILFOIE, ERN RARE‐LIVER, Centre de recherche sur l'inflammationParisFrance
| | - Jordi Gracia‐Sancho
- Liver Vascular Biology Research GroupIDIBAPS Biomedical Research Institute & CIBEREHDBarcelonaSpain,Department of Visceral Surgery and Medicine & Department for Biomedical ResearchInselspital, University of BernBernSwitzerland
| |
Collapse
|
35
|
Cizkova K, Koubova K, Tauber Z. Lipid Messenger Phosphatidylinositol-4,5-Bisphosphate Is Increased by Both PPARα Activators and Inhibitors: Relevance for Intestinal Cell Differentiation. BIOLOGY 2022; 11:biology11070997. [PMID: 36101378 PMCID: PMC9312331 DOI: 10.3390/biology11070997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 11/16/2022]
Abstract
Simple Summary Fibrates, such as fenofibrate, are widely used drugs for dyslipidaemia treatment. It is known that they activate peroxisome proliferator-activated receptor α (PPARα) which serves as a lipid sensor in the organism. This article addresses how activators and inhibitor of the PPARα could affect differentiation of intestinal cells. Carcinogenesis is a disruption of normal differentiation process and colorectal carcinoma is the third most common cancer in terms of incidence, but the secondp in terms of mortality. One of the important signalling pathways in intestinal cell differentiation as well as carcinogenesis is PI3K/Akt/PTEN. We showed that PPARα activators as well as inhibitor affected the levels of one member of this pathway called phosphatidylinositol-4,5-bisphosphate. This molecule is important for formation of microvilli, the essential structures of fully differentiated intestinal cells. Abstract We investigated the effects of PPARα activators fenofibrate and WY-14643 as well as the PPARα inhibitor GW6471 on the PI3K/Akt/PTEN pathway of intestinal cell differentiation. Our previous study showed that all these compounds increased the expression of villin, a specific marker of intestinal cell differentiation in HT-29 and Caco2 cells. Our current results confirmed the central role of lipid messenger phosphatidylinositol-4,5-bisphosphate (PIP2), a known player in brush border formation, in mediating the effects of tested PPARα ligands. Although all tested compounds increased its levels, surprisingly, each of them affected different PIP2-metabolizing enzymes, especially the levels of PIP5K1C and PTEN. Moreover, we found a positive relationship between the expression of PPARα itself and PIP2 as well as PIP5K1C. By contrast, PPARα was negatively correlated with PTEN. However, the expression of antigens of interest was independent of PPARα subcellular localization, suggesting that it is not directly involved in their regulation. In colorectal carcinoma tissues we found a decrease in PTEN expression, which was accompanied by a change in its subcellular localization. This change was also observed for the regulatory subunit of PI3K. Taken together, our data revealed that fenofibrate, WY-14643, and GW6471 affected different members of the PI3K/Akt/PTEN pathway. However, these effects were PPARα-independent.
Collapse
|
36
|
Setz C, Große M, Auth J, Fröba M, Rauch P, Bausch A, Wright M, Schubert U. Synergistic Antiviral Activity of Pamapimod and Pioglitazone against SARS-CoV-2 and Its Variants of Concern. Int J Mol Sci 2022; 23:6830. [PMID: 35743273 PMCID: PMC9224751 DOI: 10.3390/ijms23126830] [Citation(s) in RCA: 7] [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: 05/05/2022] [Revised: 06/08/2022] [Accepted: 06/10/2022] [Indexed: 11/17/2022] Open
Abstract
The SARS-CoV-2 pandemic remains a major public health threat, especially due to newly emerging SARS-CoV-2 Variants of Concern (VoCs), which are more efficiently transmitted, more virulent, and more able to escape naturally acquired and vaccine-induced immunity. Recently, the protease inhibitor Paxlovid® and the polymerase inhibitor molnupiravir, both targeting mutant-prone viral components, were approved for high-risk COVID-19 patients. Nevertheless, effective therapeutics to treat COVID-19 are urgently needed, especially small molecules acting independently of VoCs and targeting genetically stable cellular pathways which are crucial for viral replication. Pamapimod is a selective inhibitor of p38 Mitogen-Activated Protein Kinase alpha (p38 MAPKα) that has been extensively clinically evaluated for the treatment of rheumatoid arthritis. Signaling via p38 has recently been described as a key pathway for the replication of SARS-CoV-2. Here, we reveal that the combination of pamapimod with pioglitazone, an anti-inflammatory and approved drug for the treatment of type 2 diabetes, possesses potent and synergistic activity to inhibit SARS-CoV-2 replication in vitro. Both drugs showed similar antiviral potency across several cultured cell types and similar antiviral activity against SARS-CoV-2 Wuhan type, and the VoCs Alpha, Beta, Gamma, Delta, and Omicron. These data support the combination of pamapimod and pioglitazone as a potential therapy to reduce duration and severity of disease in COVID-19 patients, an assumption currently evaluated in an ongoing phase II clinical study.
Collapse
Affiliation(s)
- Christian Setz
- Institute of Virology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (C.S.); (M.G.); (J.A.); (M.F.); (P.R.)
| | - Maximilian Große
- Institute of Virology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (C.S.); (M.G.); (J.A.); (M.F.); (P.R.)
| | - Janina Auth
- Institute of Virology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (C.S.); (M.G.); (J.A.); (M.F.); (P.R.)
| | - Maria Fröba
- Institute of Virology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (C.S.); (M.G.); (J.A.); (M.F.); (P.R.)
| | - Pia Rauch
- Institute of Virology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (C.S.); (M.G.); (J.A.); (M.F.); (P.R.)
| | | | | | - Ulrich Schubert
- Institute of Virology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (C.S.); (M.G.); (J.A.); (M.F.); (P.R.)
| |
Collapse
|
37
|
Cai Q, Song Y, Wang S, Wang W, Sun X, Yu J, Wei Y. Functional yogurt fermented by two-probiotics regulates blood lipid and weight in a high-fat diet mouse model. J Food Biochem 2022; 46:e14248. [PMID: 35638246 DOI: 10.1111/jfbc.14248] [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: 01/04/2022] [Revised: 04/19/2022] [Accepted: 05/04/2022] [Indexed: 11/30/2022]
Abstract
We investigated the blood lipid regulation effects and mechanism of a functional Natto yogurt in a high-fat diet-induced hyperlipidemia mouse model. Natto yogurt was characteristically fermented by Bacillus natto and Lactobacillus plantarum with milk-soy dual protein as substrates. After 5 weeks of Natto yogurt consumption, the body weight, fat, and liver weight of mice were significantly improved, while serum levels of TG, TC, LDL, ALT, TBIL, and TBA were reduced. Natto yogurt significantly decreased the area of liver fat infiltration and the number of lipid droplets. In mechanism, we found that Natto yogurt can inhibit fatty acid synthesis and enhance fatty acid catabolism by regulating the expression of PPARα, PPARγ, CD36 and FAS in the liver. Moreover, Natto yogurt increased the ratio of Bacteroidetes to Firmicutes in the intestine. These results provide a possibility for Natto yogurt as a dual protein functional food to prevent and treat hyperlipidemia and obesity. PRACTICAL APPLICATIONS: Traditional-fermented yogurt promotes nutritional absorption and reduces blood pressure and fat, while Bacillus natto and its fermented food have been proved to play a significant role in improving cardiovascular and cerebrovascular diseases and obesity. Therefore, we developed a new dual protein functional yogurt (Natto yogurt) fermented by B. natto and Lactobacillus plantarum with milk and soy as substrates. We found that Natto yogurt could notably regulate blood lipid by inhibiting the synthesis of fatty acids, accelerating the catabolism of fatty acids, reducing liver damage, and increasing the abundance of beneficial intestinal microorganisms. This study suggested that Natto yogurt could improve hyperlipidemia and obesity as a safe, effective, and healthy functional food.
Collapse
Affiliation(s)
- Qinling Cai
- College of Life Sciences, Qingdao University, Qingdao, China
| | - Yahui Song
- College of Life Sciences, Qingdao University, Qingdao, China
| | - Shanglong Wang
- Chenland Nutritionals, Incorporated, Invine, California, USA
| | - Weihong Wang
- Haisenbao (Yantai) Biotechnology Development Co., Ltd, Yantai, China
| | - Xiaopeng Sun
- College of Life Sciences, Qingdao University, Qingdao, China
| | - Jia Yu
- College of Life Sciences, Qingdao University, Qingdao, China
| | - Yuxi Wei
- College of Life Sciences, Qingdao University, Qingdao, China
| |
Collapse
|
38
|
Bécsi B, Kónya Z, Boratkó A, Kovács K, Erdődi F. Epigallocatechine-3-gallate Inhibits the Adipogenesis of Human Mesenchymal Stem Cells via the Regulation of Protein Phosphatase-2A and Myosin Phosphatase. Cells 2022; 11:cells11101704. [PMID: 35626740 PMCID: PMC9140100 DOI: 10.3390/cells11101704] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 02/01/2023] Open
Abstract
Epigallocatechin-3-gallate (EGCG) has widespread effects on adipocyte development. However, the molecular mechanisms of EGCG are not fully understood. We investigate the adipogenic differentiation of human-derived mesenchymal stem cells, including lipid deposition and changes in the expression and phosphorylation of key transcription factors, myosin, protein phosphatase-2A (PP2A), and myosin phosphatase (MP). On day 6 of adipogenic differentiation, EGCG (1–20 µM) suppressed lipid droplet formation, which was counteracted by an EGCG-binding peptide for the 67 kDa laminin receptor (67LR), suggesting that EGCG acts via 67LR. EGCG decreased the phosphorylation of CCAAT-enhancer-binding protein beta via the activation of PP2A in a protein kinase A (PKA)-dependent manner, leading to the partial suppression of peroxisome proliferator-activated receptor gamma (PPARγ) and adiponectin expression. Differentiated cells exhibited a rounded shape, cortical actin filaments, and lipid accumulation. The EGCG treatment induced cell elongation, stress fiber formation, and less lipid accumulation. These effects were accompanied by the degradation of the MP target subunit-1 and increased the phosphorylation of the 20 kDa myosin light chain. Our results suggest that EGCG acts as an agonist of 67LR to inhibit adipogenesis via the activation of PP2A and suppression of MP. These events are coupled with the decreased phosphorylation and expression levels of adipogenic transcription factors and changes in cell shape, culminating in curtailed adipogenesis.
Collapse
Affiliation(s)
- Bálint Bécsi
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (B.B.); (Z.K.); (A.B.); (K.K.)
| | - Zoltán Kónya
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (B.B.); (Z.K.); (A.B.); (K.K.)
- MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Anita Boratkó
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (B.B.); (Z.K.); (A.B.); (K.K.)
| | - Katalin Kovács
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (B.B.); (Z.K.); (A.B.); (K.K.)
- MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Ferenc Erdődi
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (B.B.); (Z.K.); (A.B.); (K.K.)
- MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
- Correspondence: ; Tel.: +36-52-412345
| |
Collapse
|
39
|
Zhao Y, Castro LFC, Monroig Ó, Cao X, Sun Y, Gao J. A zebrafish pparγ gene deletion reveals a protein kinase network associated with defective lipid metabolism. Funct Integr Genomics 2022; 22:435-450. [PMID: 35290539 DOI: 10.1007/s10142-022-00839-7] [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: 09/07/2021] [Revised: 02/26/2022] [Accepted: 02/28/2022] [Indexed: 11/25/2022]
Abstract
Peroxisome proliferator-activated receptor γ (Pparγ) is a master regulator of adipogenesis. Chronic pathologies such as obesity, cardiovascular diseases, and diabetes involve the dysfunction of this transcription factor. Here, we generated a zebrafish mutant in pparγ (KO) with CRISPR/Cas9 technology and revealed its regulatory network. We uncovered the hepatic phenotypes of these male and female KO, and then the male wild-type zebrafish (WT) and KO were fed with a high-fat (HF) or standard diet (SD). We next conducted an integrated analyze of the proteomics and phosphoproteomics profiles. Compared with WT, the KO showed remarkable hyalinization and congestion lesions in the liver of males. Strikingly, pparγ deletion protected against the influence of high-fat diet feeding on lipid deposition in zebrafish. Some protein kinases critical for lipid metabolism, including serine/threonine-protein kinase TOR (mTOR), ribosomal protein S6 kinase (Rps6kb1b), and mitogen-activated protein kinase 14A (Mapk14a), were identified to be highly phosphorylated in KO based on differential proteome and phosphoproteome analysis. Our study supplies a pparγ deletion animal model and provides a comprehensive description of pparγ-induced expression level alterations of proteins and their phosphorylation, which are vital to understand the defective lipid metabolism risks posed to human health.
Collapse
Affiliation(s)
- Yan Zhao
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
- Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, 430070, China
- School of Basic Medical Sciences, Wuhan University, Wuhan, 430070, China
| | - L Filipe C Castro
- CIIMAR/CIMAR - Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Porto, Portugal
- FCUP - Faculty of Sciences, Department of Biology, University of Porto, Porto, Portugal
| | - Óscar Monroig
- Instituto de Acuicultura Torre de La Sal (IATS-CSIC), Ribera de Cabanes, 12595, Castellón, Spain
| | - Xiaojuan Cao
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
- Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, 430070, China
| | - Yonghua Sun
- State Key Laboratory of Freshwater Ecology and Biotechnology, China Zebrafish Resource Center, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430070, China
| | - Jian Gao
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China.
- Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, 430070, China.
| |
Collapse
|
40
|
Kamaruddin NN, Mohd Din LH, Jack A, Abdul Manan AF, Mohamad H, Tengku Muhammad TS. Acanthaster planci Inhibits PCSK9 Gene Expression via Peroxisome Proliferator Response Element (PPRE) and Activation of MEK and PKC Signaling Pathways in Human Liver Cells. Pharmaceuticals (Basel) 2022; 15:ph15030269. [PMID: 35337067 PMCID: PMC8955981 DOI: 10.3390/ph15030269] [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: 12/17/2021] [Revised: 02/13/2022] [Accepted: 02/14/2022] [Indexed: 02/04/2023] Open
Abstract
A constantly elevated level of low-density lipoprotein cholesterol (LDL-C) is mainly associated with the development of atherosclerosis. The use of statins as a treatment for reducing plasma LDL-C levels has led, in some cases, to adverse side effects, including a decrease in hepatic LDL receptor (LDLR), the receptor responsible for the uptake of circulating LDL-C. Proprotein convertase subtilisin/kexin type 9 (PCSK9) is an enzyme responsible for directing the LDLR–LDL-C complex to lysosomal degradation upon transport into cells, preventing the recycling of LDLR to the cell surface. Therefore, PCSK9 may offer a new target for reducing the levels of plasma LDL-C. In this study, we investigated the mechanisms of action of a selected fraction of A. planci on PCSK9 gene expression, as well as the effect of the fraction on the level of LDLR protein and the uptake of LDL-C. Using real-time PCR, it was shown that the selected A. planci fraction reduced the gene expression of PCSK9 in human liver HepG2 cells. Immunocytochemistry analysis demonstrated that the selected A. planci fraction increased the LDLR protein level and LDL-C uptake in HepG2 cells. Promoter mutational and gene expression analyses revealed that PPRE, a binding site for peroxisome proliferator–activated receptor (PPAR), was responsible for mediating the inhibitory effect of the selected fraction on PCSK9 mRNA. In addition, MAP kinase and PKC components of the signal transduction pathway were activated, inducing the action of the selected A. planci fraction in decreasing PCSK9 gene expression. These findings suggest that the selected fraction shows good potential for reducing circulating LDL-C and, thus, may be a good therapeutic intervention to prevent the progression of atherosclerosis.
Collapse
Affiliation(s)
- Nurjannatul Naim Kamaruddin
- Immune and Molecular Therapeutics Program, Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Kuala Terengganu 21030, Terengganu, Malaysia; (N.N.K.); (L.H.M.D.); (A.J.)
| | - Lukman Hakim Mohd Din
- Immune and Molecular Therapeutics Program, Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Kuala Terengganu 21030, Terengganu, Malaysia; (N.N.K.); (L.H.M.D.); (A.J.)
| | - Allicia Jack
- Immune and Molecular Therapeutics Program, Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Kuala Terengganu 21030, Terengganu, Malaysia; (N.N.K.); (L.H.M.D.); (A.J.)
- Nutrition & Food Safety Program, Food Science & Technology Research Centre, Malaysian Agricultural Research & Development Institute (MARDI) Headquarters, Serdang 43400, Selangor, Malaysia
| | - Aina Farahiyah Abdul Manan
- Natural and Product Synthetics Program, Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Kuala Terengganu 21030, Terengganu, Malaysia; (A.F.A.M.); (H.M.)
| | - Habsah Mohamad
- Natural and Product Synthetics Program, Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Kuala Terengganu 21030, Terengganu, Malaysia; (A.F.A.M.); (H.M.)
| | - Tengku Sifzizul Tengku Muhammad
- Immune and Molecular Therapeutics Program, Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Kuala Terengganu 21030, Terengganu, Malaysia; (N.N.K.); (L.H.M.D.); (A.J.)
- Correspondence:
| |
Collapse
|
41
|
Hanaguri J, Nagai N, Yokota H, Kushiyama A, Watanabe M, Yamagami S, Nagaoka T. Fenofibrate Nano-Eyedrops Ameliorate Retinal Blood Flow Dysregulation and Neurovascular Coupling in Type 2 Diabetic Mice. Pharmaceutics 2022; 14:pharmaceutics14020384. [PMID: 35214116 PMCID: PMC8876509 DOI: 10.3390/pharmaceutics14020384] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/22/2022] [Accepted: 01/26/2022] [Indexed: 02/05/2023] Open
Abstract
We investigated the effect of fenofibrate nano-eyedrops (FenoNano) on impaired retinal blood flow regulation in type 2 diabetic mice. Six-week-old db/db mice were randomly divided into an untreated group (n = 6) and treated group, which received FenoNano (n = 6). The longitudinal changes in retinal neuronal function and blood flow responses to systemic hyperoxia and flicker stimulation were evaluated every 2 weeks in diabetic db/db mice treated with FenoNano (n = 6) or the vehicle (n = 6) from ages 8–14 weeks. The retinal blood flow was assessed using laser speckle flowgraphy. We also evaluated the expressions of vascular endothelial growth factor (VEGF), glial fibrillary acidic protein (GFAP), and aquaporin 4 (AQP4) and the phosphorylation of peroxisome proliferator-activated receptor alpha (PPAR-α) by immunofluorescence. In db/db mice treated with FenoNano, both responses were restored from 8 to 14 weeks of age compared with the diabetic mice treated with the vehicle. At 14 weeks of age, the impaired regulation of retinal blood flow during systemic hyperoxia and flicker stimulation improved to about half of that in the db/db mice treated with FenoNano compared with the db/m control group (n = 5). FenoNano prevented the activation of VEGF and GFAP expression and increased the AQP4 expression and the phosphorylation of PPAR-α detected by immunofluorescence compared with the diabetic mice treated with the vehicle eyedrop. Our results suggested that the fenofibrate nano-eyedrops prevent retinal glial dysfunction via the phosphorylation of PPAR-α and improves the retinal blood flow dysregulation in type 2 diabetic mice.
Collapse
Affiliation(s)
- Junya Hanaguri
- Department of Visual Science, Division of Ophthalmology, Nihon University School of Medicine, Itabashi-ku, Tokyo 173-8610, Japan; (J.H.); (H.Y.); (M.W.); (S.Y.)
| | - Noriaki Nagai
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Kindai University, Higashi-Osaka 577-8502, Osaka, Japan;
| | - Harumasa Yokota
- Department of Visual Science, Division of Ophthalmology, Nihon University School of Medicine, Itabashi-ku, Tokyo 173-8610, Japan; (J.H.); (H.Y.); (M.W.); (S.Y.)
| | - Akifumi Kushiyama
- Department of Pharmacotherapy, Meiji Pharmaceutical University, Kiyose, Tokyo 204-8588, Japan;
| | - Masahisa Watanabe
- Department of Visual Science, Division of Ophthalmology, Nihon University School of Medicine, Itabashi-ku, Tokyo 173-8610, Japan; (J.H.); (H.Y.); (M.W.); (S.Y.)
| | - Satoru Yamagami
- Department of Visual Science, Division of Ophthalmology, Nihon University School of Medicine, Itabashi-ku, Tokyo 173-8610, Japan; (J.H.); (H.Y.); (M.W.); (S.Y.)
| | - Taiji Nagaoka
- Department of Visual Science, Division of Ophthalmology, Nihon University School of Medicine, Itabashi-ku, Tokyo 173-8610, Japan; (J.H.); (H.Y.); (M.W.); (S.Y.)
- Correspondence: or ; Tel.: +81-3-3972-8111
| |
Collapse
|
42
|
Differential Effects of EPA and DHA on PPARγ-mediated Sympathetic Innervation in Infarcted Rat Hearts by GPR120-dependent and -independent Mechanisms. J Nutr Biochem 2022; 103:108950. [PMID: 35121022 DOI: 10.1016/j.jnutbio.2022.108950] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 10/29/2021] [Accepted: 01/04/2022] [Indexed: 11/22/2022]
Abstract
The ω-3 fatty acids, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), have been shown to attenuate inflammation processes, whereas, the molecular mechanisms remain unclear. This study was aimed at figuring out the differential effects of EPA and DHA on fatal arrhythmias and whether the signaling pathway could be a target after myocardial infarction, an inflammatory status. Male Wistar rats after ligating coronary artery were randomized to either vehicle, EPA, or DHA for 4 weeks. Postinfarction was associated with increased myocardial norepinephrine levels and sympathetic innervation. Furthermore, infarction was associated with the activation of NLRP3 inflammasomes and increased the protein and expression of IL-1β and nerve growth factor (NGF). These changes were blunted after adding either EPA or DHA with a greater extent of EPA than DHA. Immunoblotting and immunohistochemical analysis showed that EPA had significantly lower phosphorylation of PPARγ at Ser 112 compared with DHA. Arrhythmic severity during programmed stimulation in the infarcted rats treated with EPA was significantly lower than those treated with DHA. Specific inhibition of GPR120 by AH-7614 and PPARγ by T0070907 reduced the EPA-or DHA-related attenuation of IL-1β and NGF release. Besides, AH-7614 treatment partially reduced the PPARγ levels, whereas, T0070907 administration did not affect the GPR120 levels. These results suggest that EPA was more effective than DHA in prevention of fatal arrhythmias by inhibiting NLRP3 inflammasome and sympathetic innervation through activation of PPARγ-mediated GPR120-dependent and -independent signaling pathways in infarcted hearts.
Collapse
|
43
|
Yang Y, Wang Y, Zhang Z, Wang S, Li Z. WSF-CT-11, a Sesquiterpene Derivative, Activates AMP-Activated Protein Kinase with Anti-diabetic Effects in 3T3-L1 Adipocytes. ACS OMEGA 2021; 6:31272-31281. [PMID: 34841171 PMCID: PMC8613856 DOI: 10.1021/acsomega.1c05061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/18/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND AMP-activated protein kinase (AMPK) is a therapeutic target against type II diabetes (T2D). Synthetic sesquiterpene derivatives were investigated to identify novel AMPK activators as anti-diabetic drugs because the leading drugs like metformin and thiazolidinediones (TZDs) activate AMPK by inhibiting the synthesis of adenosine 5'-triphosphate and thus are associated with some side effects. RESULTS We identified WSF-CT-11 as an AMPK activator in HEK293T cells and found that WSF-CT-11 activates AMPK by the activation of transient receptor potential vanilloid type 1 (TRPV1), a Ca2+-permeable cation channel. The increased Ca2+ influx then activates phosphoinositide 3-kinase (PI3K), protein kinase B (PKB/Akt), and Ca2+/calmodulin-dependent protein kinase II (CaMKII), which in turn phosphorylates TRPV1 and facilitates the formation of a TRPV1/Akt/CaMKII/AMPK complex. This complex might be important for the regulation of AMPK activity. In 3T3-L1 adipocytes, WSF-CT-11-induced AMPK activation has three anti-diabetic effects. It promotes the assembly of high-molecular-weight adiponectin, which has stronger insulin-sensitizing activity than other multimers. It improves translocation of the glucose transporter type 4, increases glucose uptake, and induces the inhibitory phosphorylation of peroxisome proliferator-activated receptor γ and thus suppresses adipogenesis. CONCLUSION WSF-CT-11 is a novel AMPK activator and potential drug against T2D without the side effects of metformin and TZDs.
Collapse
Affiliation(s)
- Yang Yang
- MOE
Key Laboratory of Bioinoformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yunyun Wang
- College
of Chemical Engineering, Nanjing Forestry
University, Jiangsu 210037, China
| | - Zhijie Zhang
- Institute
of Chinese Materia Medica, China Academy
of Chinese Medical Sciences, Beijing 100700, China
| | - Shifa Wang
- College
of Chemical Engineering, Nanjing Forestry
University, Jiangsu 210037, China
| | - Zhen Li
- MOE
Key Laboratory of Bioinoformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
44
|
He L, Jhong JH, Chen Q, Huang KY, Strittmatter K, Kreuzer J, DeRan M, Wu X, Lee TY, Slavov N, Haas W, Marneros AG. Global characterization of macrophage polarization mechanisms and identification of M2-type polarization inhibitors. Cell Rep 2021; 37:109955. [PMID: 34731634 PMCID: PMC8783961 DOI: 10.1016/j.celrep.2021.109955] [Citation(s) in RCA: 167] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/20/2021] [Accepted: 10/15/2021] [Indexed: 01/07/2023] Open
Abstract
Macrophages undergoing M1- versus M2-type polarization differ significantly in their cell metabolism and cellular functions. Here, global quantitative time-course proteomics and phosphoproteomics paired with transcriptomics provide a comprehensive characterization of temporal changes in cell metabolism, cellular functions, and signaling pathways that occur during the induction phase of M1- versus M2-type polarization. Significant differences in, especially, metabolic pathways are observed, including changes in glucose metabolism, glycosaminoglycan metabolism, and retinoic acid signaling. Kinase-enrichment analysis shows activation patterns of specific kinases that are distinct in M1- versus M2-type polarization. M2-type polarization inhibitor drug screens identify drugs that selectively block M2- but not M1-type polarization, including mitogen-activated protein kinase kinase (MEK) and histone deacetylase (HDAC) inhibitors. These datasets provide a comprehensive resource to identify specific signaling and metabolic pathways that are critical for macrophage polarization. In a proof-of-principle approach, we use these datasets to show that MEK signaling is required for M2-type polarization by promoting peroxisome proliferator-activated receptor-γ (PPARγ)-induced retinoic acid signaling.
Collapse
Affiliation(s)
- Lizhi He
- Cutaneous Biology Research Center, Massachusetts General Hospital, and Department of Dermatology, Harvard Medical School, Charlestown, MA 02129, USA
| | - Jhih-Hua Jhong
- Department of Computer Science and Engineering, Yuan Ze University, Taoyuan 320, Taiwan; Warshel Institute for Computational Biology, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Qi Chen
- Warshel Institute for Computational Biology, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Kai-Yao Huang
- Department of Medical Research, Hsinchu Mackay Memorial Hospital, Hsinchu 300, Taiwan
| | - Karin Strittmatter
- Cutaneous Biology Research Center, Massachusetts General Hospital, and Department of Dermatology, Harvard Medical School, Charlestown, MA 02129, USA
| | - Johannes Kreuzer
- Cancer Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Michael DeRan
- Cutaneous Biology Research Center, Massachusetts General Hospital, and Department of Dermatology, Harvard Medical School, Charlestown, MA 02129, USA
| | - Xu Wu
- Cutaneous Biology Research Center, Massachusetts General Hospital, and Department of Dermatology, Harvard Medical School, Charlestown, MA 02129, USA
| | - Tzong-Yi Lee
- Warshel Institute for Computational Biology, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Nikolai Slavov
- Department of Bioengineering and Department of Biology, Northeastern University, Boston, MA 02115, USA
| | - Wilhelm Haas
- Cancer Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Alexander G Marneros
- Cutaneous Biology Research Center, Massachusetts General Hospital, and Department of Dermatology, Harvard Medical School, Charlestown, MA 02129, USA.
| |
Collapse
|
45
|
Kotlyarov S, Bulgakov A. Lipid Metabolism Disorders in the Comorbid Course of Nonalcoholic Fatty Liver Disease and Chronic Obstructive Pulmonary Disease. Cells 2021; 10:2978. [PMID: 34831201 PMCID: PMC8616072 DOI: 10.3390/cells10112978] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/25/2021] [Accepted: 10/30/2021] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is currently among the most common liver diseases. Unfavorable data on the epidemiology of metabolic syndrome and obesity have increased the attention of clinicians and researchers to the problem of NAFLD. The research results allow us to emphasize the systemicity and multifactoriality of the pathogenesis of liver parenchyma lesion. At the same time, many aspects of its classification, etiology, and pathogenesis remain controversial. Local and systemic metabolic disorders are also a part of the pathogenesis of chronic obstructive pulmonary disease and can influence its course. The present article analyzes the metabolic pathways mediating the links of impaired lipid metabolism in NAFLD and chronic obstructive pulmonary disease (COPD). Free fatty acids, cholesterol, and ceramides are involved in key metabolic and inflammatory pathways underlying the pathogenesis of both diseases. Moreover, inflammation and lipid metabolism demonstrate close links in the comorbid course of NAFLD and COPD.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia;
| | | |
Collapse
|
46
|
Li W, Deng M, Gong J, Zhang X, Ge S, Zhao L. Sodium Acetate Inhibit TGF-β1-Induced Activation of Hepatic Stellate Cells by Restoring AMPK or c-Jun Signaling. Front Nutr 2021; 8:729583. [PMID: 34660662 PMCID: PMC8515000 DOI: 10.3389/fnut.2021.729583] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/02/2021] [Indexed: 11/13/2022] Open
Abstract
Short-chain fatty acids (SCFAs) are crucial gut microbial metabolites that play a major role in the occurrence and development of hepatic fibrosis (HF). However, the effect of SCFAs on hepatic stellate cells (HSCs), the major pro-fibrogenic cells, is yet undefined. In this study, the effects of three major SCFAs (acetate, propionate, and butyrate) were assessed on the activation of HSCs. LX2 cells were activated with TGF-β1 and treated with sodium acetate (NaA), sodium propionate (NaP), or sodium butyrate (NaB). SCFA treatment significantly reduced the protein levels of α-SMA and the phosphorylation of Smad2 and decreased the mRNA expression of Acta2/Col1a1/Fn in cells compared to the TGF-β1 treatment. Among the three SCFAs, NaA revealed the best efficacy at alleviating TGF-β1-induced LX2 cell activation. Additionally, acetate accumulated in the cells, and G protein-coupled receptor (GPR) 43 silencing did not have any impact on the inhibition of LX2 cell activation by NaA. These findings indicated that NaA enters into the cells to inhibit LX2 cell activation independent of GPR43. The results of phosphokinase array kit and Western blot indicated that NaA increased the AMP-activated protein kinase (AMPK) activation and reduced the phosphorylation of c-Jun in cultured LX2 cells, and siRNA-peroxisome proliferator-activated receptor (PPAR) -γ abolished the inhibitory effects of NaA against TGF-β1-induced LX2 cell activation. In conclusion, this study showed that NaA inhibited LX2 cell activation by activating the AMPK/PPARγ and blocking the c-Jun signaling pathways. Thus, SCFAs might represent a novel and viable approach for alleviating HF.
Collapse
Affiliation(s)
- Weiwei Li
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Mingjuan Deng
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Jiahui Gong
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Xiaoying Zhang
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
| | - Shaoyang Ge
- Hebei Engineering Research Center of Animal Product, Sanhe, China
| | - Liang Zhao
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.,Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| |
Collapse
|
47
|
Emodin attenuates severe acute pancreatitis-associated acute lung injury by suppressing pancreatic exosome-mediated alveolar macrophage activation. Acta Pharm Sin B 2021; 12:3986-4003. [PMID: 36213542 PMCID: PMC9532455 DOI: 10.1016/j.apsb.2021.10.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/30/2021] [Accepted: 10/03/2021] [Indexed: 12/23/2022] Open
Abstract
Severe acute pancreatitis-associated acute lung injury (SAP-ALI) is a serious disease associated with high mortality. Emodin has been applied to alleviate SAP-ALI; however, the mechanism remains unclear. We report that the therapeutic role of emodin in attenuating SAP-ALI is partly dependent on an exosomal mechanism. SAP rats had increased levels of plasma exosomes with altered protein contents compared to the sham rats. These infused plasma exosomes tended to accumulate in the lungs and promoted the hyper-activation of alveolar macrophages and inflammatory damage. Conversely, emodin treatment decreased the plasma/pancreatic exosome levels in the SAP rats. Emodin-primed exosomes showed less pro-inflammatory effects in alveolar macrophages and lung tissues than SAP exosomes. In detail, emodin-primed exosomes suppressed the NF-κB pathway to reduce the activation of alveolar macrophage and ameliorate lung inflammation by regulating PPARγ pathway, while these effects were amplified/abolished by PPARγ agonist/antagonist. Blockage of pancreatic acinar cell exosome biogenesis also exhibited suppression of alveolar macrophage activation and reduction of lung inflammation. This study suggests a vital role of exosomes in participating inflammation-associated organ-injury, and indicates emodin can attenuate SAP-ALI by reducing the pancreatic exosome-mediated alveolar macrophage activation.
Collapse
|
48
|
Ng SP, Nomura W, Takahashi H, Inoue K, Kawada T, Goto T. Methylglyoxal attenuates isoproterenol-induced increase in uncoupling protein 1 expression through activation of JNK signaling pathway in beige adipocytes. Biochem Biophys Rep 2021; 28:101127. [PMID: 34527816 PMCID: PMC8430270 DOI: 10.1016/j.bbrep.2021.101127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/27/2021] [Accepted: 09/01/2021] [Indexed: 11/17/2022] Open
Abstract
Methylglyoxal (MG) is a metabolite derived from glycolysis whose levels in the blood and tissues of patients with diabetes are higher than those of healthy individuals, suggesting that MG is associated with the development of diabetic complications. However, it remains unknown whether high levels of MG are a cause or consequence of diabetes. Here, we show that MG negatively affects the expression of uncoupling protein 1 (UCP1), which is involved in thermogenesis and the regulation of systemic metabolism. Decreased Ucp1 expression is associated with obesity and type 2 diabetes. We found that MG attenuated the increase in Ucp1 expression following treatment with isoproterenol in beige adipocytes. However, MG did not affect protein kinase A signaling, the core coordinator of isoproterenol-induced Ucp1 expression. Instead, MG activated c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinases. We found that JNK inhibition, but not p38, recovered isoproterenol-stimulated Ucp1 expression under MG treatment. Altogether, these results suggest an inhibitory role of MG on the thermogenic function of beige adipocytes through the JNK signaling pathway.
Collapse
Key Words
- BBGC, S-p-bromobenzylglutathione cyclopentyl diester
- Beige adipocytes
- CREB, cAMP response element-binding protein
- ERK, extracellular receptor kinase
- HSL, hormone-sensitive lipase
- JNK
- JNK, c-Jun N-terminal kinase
- MG, methylglyoxal
- Methylglyoxal
- NAC, N-acetyl-l-cysteine
- NEFA, non-esterified fatty acids
- PKA, protein kinase A
- SEM, standard error of the mean
- Ucp1
- iWAT, inguinal white adipose tissue
Collapse
Affiliation(s)
- Su-Ping Ng
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Wataru Nomura
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan
- Research Unit for Physiological Chemistry, The Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8317, Japan
- Corresponding author. Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan.
| | - Haruya Takahashi
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Kazuo Inoue
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan
- Research Unit for Physiological Chemistry, The Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8317, Japan
| | - Teruo Kawada
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan
- Research Unit for Physiological Chemistry, The Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8317, Japan
| | - Tsuyoshi Goto
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan
- Research Unit for Physiological Chemistry, The Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8317, Japan
- Corresponding author. Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan.
| |
Collapse
|
49
|
Yang Z, Roth K, Agarwal M, Liu W, Petriello MC. The transcription factors CREBH, PPARa, and FOXO1 as critical hepatic mediators of diet-induced metabolic dysregulation. J Nutr Biochem 2021; 95:108633. [PMID: 33789150 PMCID: PMC8355060 DOI: 10.1016/j.jnutbio.2021.108633] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 01/31/2021] [Accepted: 03/09/2021] [Indexed: 02/06/2023]
Abstract
The liver is a critical mediator of lipid and/or glucose homeostasis and is a primary organ involved in dynamic changes during feeding and fasting. Additionally, hepatic-centric pathways are prone to dysregulation during pathophysiological states including metabolic syndrome (MetS) and non-alcoholic fatty liver disease. Omics platforms and GWAS have elucidated genes related to increased risk of developing MetS and related disorders, but mutations in these metabolism-related genes are rare and cannot fully explain the increasing prevalence of MetS-related pathologies worldwide. Complex interactions between diet, lifestyle, environmental factors, and genetic predisposition jointly determine inter-individual variability of disease risk. Given the complexity of these interactions, researchers have focused on master regulators of metabolic responses incorporating and mediating the impact of multiple environmental cues. Transcription factors are DNA binding, terminal executors of signaling pathways that modulate the cellular responses to complex metabolic stimuli and are related to the control of hepatic lipid and glucose homeostasis. Among numerous hepatic transcription factors involved in regulating metabolism, three emerge as key players in transducing nutrient sensing, which are dysregulated in MetS-related perturbations in both clinical and preclinical studies: cAMP Responsive Element Binding Protein 3 Like 3 (CREB3L3), Peroxisome Proliferator Activated Receptor Alpha (PPAR), and Forkhead Box O1 (FOXO1). Additionally, these three transcription factors appear to be amenable to dietary and/or nutrient-based therapies, being potential targets of nutritional therapy. In this review we aim to describe the activation, regulation, and impact of these transcription factors in the context of metabolic homeostasis. We also summarize their perspectives in MetS and nutritional therapies.
Collapse
Affiliation(s)
- Zhao Yang
- Institute of Environmental Health Sciences (IEHS), Wayne State University, Detroit, MI, USA
| | - Katherine Roth
- Institute of Environmental Health Sciences (IEHS), Wayne State University, Detroit, MI, USA
| | - Manisha Agarwal
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Wanqing Liu
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, USA; Department of Pharmaceutical Sciences, College of Pharmacy, Wayne State University, Detroit, MI, USA
| | - Michael C Petriello
- Institute of Environmental Health Sciences (IEHS), Wayne State University, Detroit, MI, USA; Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
50
|
Yao J, Wang Z, Wang R, Wang Y, Xu J, He X. Anti-proliferative and anti-inflammatory prenylated isoflavones and coumaronochromones from the fruits of Ficus altissima. Bioorg Chem 2021; 64:2893-900. [PMID: 34038794 DOI: 10.1021/acs.jafc.6b00227] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2023]
Abstract
Ficus altissima, an evergreen arbor belonging to the Moraceae family, is mainly cultivated in the tropics and subtropics of South and Southeast Asia with the characteristic of exuberant vitality and luxuriant foliage. In this article, four new prenylated isoflavones (1-4), along with ten previously described isoflavones (5-14) and two known prenylated coumaronochromones (15 and 16) were firstly obtained from the fruits of F. altissima. Their structures were identified by various spectroscopic techniques including specific optical rotation, HR-ESI-MS and NMR. The isolated products were evaluated for their anti-proliferative activities against three human tumor cell lines (HepG2, MCF-7 and MDA-MB-231) through MTT assay. Compounds 2, 3 and 16 exhibited obvious anti-proliferative activities against MDA-MB-231 cell line and compounds 3, 13 and 16 showed effective cytotoxic effects on HepG2 cell line in a concentration-dependent manner, as verified by the colony formation assay, cell and nucleus morphological assessment and apoptosis assay. Meanwhile, compounds 5 and 12 exhibited significant inhibition activities on NO production in LPS-stimulated RAW 264.7 cell line compared with positive control indometacin. The phytochemical investigation of the fruits of F. altissima in this study could provide the evidence for the discovery of lead compounds.
Collapse
Affiliation(s)
- Jiaming Yao
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zhe Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Ru Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yihai Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Engineering Research Center for Lead Compounds & Drug Discovery, Guangzhou 510006, China.
| | - Jingwen Xu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Engineering Research Center for Lead Compounds & Drug Discovery, Guangzhou 510006, China
| | - Xiangjiu He
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Engineering Research Center for Lead Compounds & Drug Discovery, Guangzhou 510006, China.
| |
Collapse
|