1
|
Yu F, Zhao X, Wang Q, Niu Y, Xiao P, Zhang J, Fei K, Huang Y, Liu L, Fang PH, Du X, Li W, He D, Zhang T, Li S, Yuan J. Photothermal-Responsive Soluble Microneedle Patches for Meibomian Gland Dysfunction Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413962. [PMID: 39887671 PMCID: PMC11923895 DOI: 10.1002/advs.202413962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/12/2025] [Indexed: 02/01/2025]
Abstract
Meibomian gland dysfunction (MGD) is a leading cause of evaporative dry eye disease, presenting a challenge for targeted treatment. Traditional topical ocular drug delivery methods often fail to effectively reach the meibomian glands (MGs). To address this, the study has developed a soluble microneedles (MN) patch comprising poly(vinyl alcohol), cyclodextrin modified polyacrylic acid, and new indocyanine green. This innovative MN patch facilitates the transdermal release of peroxisome proliferator-activated receptor gamma (PPAR-γ) agonists, such as rosiglitazone in response to near-infrared ray induced temperature changes. By safely optimizing temperature, the patch effectively liquefied meibum lips, thereby alleviating duct obstruction while releasing the drug. MN patches exhibit sufficient mechanical strength for effective skin penetration, and its biosafety for eyelid application has been rigorously assessed in vitro and in vivo. The therapeutic efficiency of rosiglitazone loaded MN (ROSI-MN) treatment for MGD is evaluated in high-fat mice. After three months of treatments, ROSI-MN administration significantly alleviated MGD clinical manifestations, including ocular surface damage, lipid deposits, glandular hypertrophy, and inflammatory infiltration, ultimately improving the microstructure and biofunction of MGs. In conclusion, the soluble MN patches hold promise as an effective drug delivery strategy for treating ocular surface diseases beyond MGD.
Collapse
Affiliation(s)
- Fei Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510623, China
- Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510020, China
| | - Xuan Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510623, China
| | - Qian Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510623, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Yifei Niu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510623, China
| | - Peng Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510623, China
| | - Jinze Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510623, China
| | - Keyi Fei
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510623, China
| | - Yuancong Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510623, China
| | - Liu Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510623, China
| | - Po-Han Fang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510623, China
| | - Xinyue Du
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510623, China
| | - Weihua Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510623, China
| | - Dalian He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510623, China
| | - Tingting Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510623, China
| | - Saiqun Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510623, China
| | - Jin Yuan
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology & Visual Sciences, Beijing, 100730, China
| |
Collapse
|
2
|
Li WR, Zhang C, Wang J. PPARs: modulating lipotoxicity and thus inhibiting fibrosis. Hormones (Athens) 2025; 24:85-97. [PMID: 39500811 DOI: 10.1007/s42000-024-00612-4] [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: 07/16/2024] [Accepted: 10/11/2024] [Indexed: 03/18/2025]
Abstract
Peroxisome proliferator-activated receptors (PPARs) belong to the nuclear hormone receptor family of ligand-activated receptors and are known for their roles as key factors in the regulation of lipid metabolism. In the more than three decades since their discovery, most reports on PPARs have focused on their roles in lipid metabolism, and a portion of the new research has also focused on the relationship between PPARs and fibrosis. Interestingly, lipid metabolism disorders and fibrosis are also inextricably linked. This implies that PPARs, lipid metabolism and fibrosis are interrelated. On this basis, we have summarized the molecular mechanisms of PPARs regulating fibrosis through lipid metabolism and PPARγ directly regulating fibrosis, and pointed out the contradictions and enigmas that need to be further explored in the processes of PPARs regulating lipid metabolism and fibrosis. The aim of the present review is to provide new ideas for PPARs for the treatment of lipid metabolism disorders and fibrosis.
Collapse
Affiliation(s)
- Wen-Rui Li
- The Second Clinical Medical College, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Chunping Zhang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Jing Wang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
3
|
Alotaiq N, Khalifa AS, Youssef A, El-Nagar EG, Elwali NE, Habib HM, AlZaim I, Eid AH, Bakkar NMZ, El-Yazbi AF. Targeting GSK-3β for adipose dysfunction and cardiovascular complications of metabolic disease: An entangled WNT/β-catenin question. FASEB J 2024; 38:e70273. [PMID: 39726401 DOI: 10.1096/fj.202402470r] [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: 10/15/2024] [Revised: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
Individuals with metabolic syndrome have a high risk of developing cardiovascular disorders that is closely tied to visceral adipose tissue dysfunction, as well as an altered interaction between adipose tissue and the cardiovascular system. In metabolic syndrome, adipose tissue dysfunction is associated with increased hypertrophy, reduced vascularization, and hypoxia of adipocytes, leading to a pro-oxidative and pro-inflammatory environment. Among the pathways regulating adipose tissue homeostasis is the wingless-type mammary tumor virus integration site family (Wnt) signaling pathway, with both its canonical and non-canonical arms. Various modulators of the Wnt signaling have been identified to contribute to the development of metabolic diseases and their cardiovascular complications, with a particularly significant role played by Glycogen Synthase Kinase-3β (GSK-3β). GSK-3β levels and activities have various and often contrasting roles in obesity and related metabolic disorders, as well as their cardiovascular sequelae. Here, we explore the possibility that altered Wnt signaling and GSK-3β activities could serve as a connection between adipose tissue dysfunction and the development of cardiovascular disease in individuals with metabolic syndrome. We attempt to define a context-specific approach for intervention, which could possibly serve as a novel disease modifying therapy for the mitigation of such complications.
Collapse
Affiliation(s)
- Nasser Alotaiq
- Health Sciences Research Center, Imam Muhammad Ibn Saud Islamic University (IMISIU), Riyadh, Kingdom of Saudi Arabia
| | - Ahmed S Khalifa
- Faculty of Pharmacy, Alamein International University, Alamein, Egypt
| | - Amr Youssef
- Faculty of Pharmacy, Alamein International University, Alamein, Egypt
| | - Esraa G El-Nagar
- Faculty of Pharmacy, Alamein International University, Alamein, Egypt
| | - Nasr Eldin Elwali
- Deanship of Scientific Research, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Kingdom of Saudi Arabia
| | - Hosam M Habib
- Research & Innovation Hub, Alamein International University, Alamein, Egypt
| | - Ibrahim AlZaim
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | | | - Ahmed F El-Yazbi
- Faculty of Pharmacy, Alamein International University, Alamein, Egypt
- Research & Innovation Hub, Alamein International University, Alamein, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
4
|
Kononova YA, Tuchina TP, Babenko AY. Brown and Beige Adipose Tissue: One or Different Targets for Treatment of Obesity and Obesity-Related Metabolic Disorders? Int J Mol Sci 2024; 25:13295. [PMID: 39769065 PMCID: PMC11677471 DOI: 10.3390/ijms252413295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 11/27/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
The failure of the fight against obesity makes us turn to new goals in its treatment. Now, brown adipose tissue has attracted attention as a promising target for the treatment of obesity and associated metabolic disorders such as insulin resistance, dyslipidemia, and glucose tolerance disorders. Meanwhile, the expansion of our knowledge has led to awareness about two rather different subtypes: classic brown and beige (inducible brown) adipose tissue. These subtypes have different origin, differences in the expression of individual genes but also a lot in common. Both tissues are thermogenic, which means that, by increasing energy consumption, they can improve their balance with excess intake. Both tissues are activated in response to specific inducers (cold, beta-adrenergic receptor activation, certain food and drugs), but beige adipose tissue transdifferentiates back into white adipose tissue after the cessation of inducing action, while classic brown adipose tissue persists, but its activity decreases. In this review, we attempted to understand whether there are differences in the effects of different groups of thermogenesis-affecting drugs on these tissues. The analysis showed that this area of research is rather sparse and requires close attention in further studies.
Collapse
Affiliation(s)
- Yulia A. Kononova
- World-Class Scientific Center “Center for Personalized Medicine”, Almazov National Medical Research Centre, 197341 St. Petersburg, Russia;
| | - Taisiia P. Tuchina
- Endocrinology Department, Almazov National Medical Research Centre, 197341 St. Petersburg, Russia;
| | - Alina Yu. Babenko
- World-Class Scientific Center “Center for Personalized Medicine”, Almazov National Medical Research Centre, 197341 St. Petersburg, Russia;
| |
Collapse
|
5
|
Galigniana NM, Ruiz MC, Piwien-Pilipuk G. FK506 binding protein 51: Its role in the adipose organ and beyond. J Cell Biochem 2024; 125:e30351. [PMID: 36502528 DOI: 10.1002/jcb.30351] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/04/2022] [Accepted: 11/15/2022] [Indexed: 02/17/2024]
Abstract
There is a great body of evidence that the adipose organ plays a central role in the control not only of energy balance, but importantly, in the maintenance of metabolic homeostasis. Interest in the study of different aspects of its physiology grew in the last decades due to the pandemic of obesity and the consequences of metabolic syndrome. It was not until recently that the first evidence for the role of the high molecular weight immunophilin FK506 binding protein (FKBP) 51 in the process of adipocyte differentiation have been described. Since then, many new facets have been discovered of this stress-responsive FKBP51 as a central node for precise coordination of many cell functions, as shown for nuclear steroid receptors, autophagy, signaling pathways as Akt, p38 MAPK, and GSK3, as well as for insulin signaling and the control of glucose homeostasis. Thus, the aim of this review is to integrate and discuss the recent advances in the understanding of the many roles of FKBP51 in the adipose organ.
Collapse
Affiliation(s)
- Natalia M Galigniana
- Laboratory of Nuclear Architecture, Instituto de Biología y Medicina Experimental (IBYME)-CONICET, Buenos Aires, Argentina
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Marina C Ruiz
- Laboratory of Nuclear Architecture, Instituto de Biología y Medicina Experimental (IBYME)-CONICET, Buenos Aires, Argentina
| | - Graciela Piwien-Pilipuk
- Laboratory of Nuclear Architecture, Instituto de Biología y Medicina Experimental (IBYME)-CONICET, Buenos Aires, Argentina
| |
Collapse
|
6
|
Małodobra-Mazur M, Ołdakowska M, Dobosz T. Exploring PPAR Gamma and PPAR Alpha's Regulation Role in Metabolism via Epigenetics Mechanism. Biomolecules 2024; 14:1445. [PMID: 39595621 PMCID: PMC11591816 DOI: 10.3390/biom14111445] [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: 09/03/2024] [Revised: 10/18/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) belong to a family of nuclear receptors. To date, three types of PPARs, namely PPARα, PPARδ, and PPARγ, have been identified, demonstrating co-expression across numerous tissues. PPARγ is primarily distributed in adipose tissue, the colon, the immune system, and the retina, while PPARα is predominantly expressed in metabolic tissues such as brown adipose tissue, the liver, and the kidneys. Both PPARγ and PPARα play crucial roles in various cellular processes. Recent data suggest that the PPAR family, among other mechanisms, might also be regulated by epigenetic mechanisms. Our recent studies, alongside numerous others, have highlighted the pivotal roles of DNA methylation and histone modifications in the regulation of PPARγ and PPARα, implicating them in the deterioration of metabolic disorders via epigenetic mechanisms. This still not fully understood mechanism of regulation in the nuclear receptors family has been summarized and described in the present paper. The present review summarizes the available data on PPARγ and PPARα regulation via epigenetic mechanisms, elucidating the link between the development of metabolic disorders and the dysregulation of PPARγ and PPARα resulting from these mechanisms.
Collapse
Affiliation(s)
- Małgorzata Małodobra-Mazur
- Department of Forensic Science, Division of Molecular Techniques, Wroclaw Medical University, Sklodowskiej-Curie 52, 51-367 Wroclaw, Poland; (M.O.); (T.D.)
| | | | | |
Collapse
|
7
|
Xu X, Charrier A, Congrove S, Ockunzzi J, Buchner DA. Cell-state-dependent regulation of PPARγ signaling by the transcription factor ZBTB9 in adipocytes. J Biol Chem 2024; 300:107985. [PMID: 39542250 DOI: 10.1016/j.jbc.2024.107985] [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: 03/11/2024] [Revised: 10/25/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024] Open
Abstract
Peroxisome proliferator-activated receptor-γ (PPARγ) is a nuclear hormone receptor that is a master regulator of adipocyte differentiation and function. ZBTB9 is a widely expressed but poorly studied transcription factor that was predicted to interact with PPARγ based on large-scale protein-protein interaction experiments. In addition, genome-wide association studies (GWAS) revealed associations between ZBTB9 and BMI, T2D risk, and HbA1c levels. Here we show that Zbtb9 deficiency in mature adipocytes decreased PPARγ activity and protein level, and thus acts as a positive regulator of PPARγ signaling. In contrast, Zbtb9 deficiency in 3T3-L1 and human preadipocytes increased PPARγ levels and enhanced adipogenesis. Transcriptomic and transcription factor binding site analyses of Zbtb9 deficient preadipocytes revealed that the E2F pathway, controlled by the E2F family of transcription factors that are classically associated with cell cycle regulation, was among the most upregulated pathways. E2F1 positively regulates adipogenesis by promoting Pparg expression, independent of its cell cycle role, via direct binding to the Pparg promoter early during adipogenesis. RB phosphorylation (pRB), which regulates E2F activity, was also upregulated in Zbtb9 deficient preadipocytes. Critically, an E2F1 inhibitor blocked the effects of Zbtb9 deficiency on adipogenesis. Collectively, these results demonstrate that Zbtb9 inhibits adipogenesis as a negative regulator of Pparg expression via pRB-E2F signaling. Our findings reveal cell-state dependent roles of ZBTB9 in adipocytes, identifying a new molecule that regulates adipocyte biology as both a positive and negative regulator of PPARγ signaling depending on the cellular context, and thus may be important in the pathogenesis of obesity and T2D.
Collapse
Affiliation(s)
- Xuan Xu
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Alyssa Charrier
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Sunny Congrove
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Jeremiah Ockunzzi
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - David A Buchner
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA; Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA.
| |
Collapse
|
8
|
Umetsu A, Watanabe M, Sato T, Higashide M, Nishikiori N, Furuhashi M, Ohguro H. TGF-β effects on adipogenesis of 3T3-L1 cells differ in 2D and 3D cell culture conditions. FEBS Open Bio 2024. [PMID: 39380256 DOI: 10.1002/2211-5463.13890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/04/2024] [Accepted: 08/23/2024] [Indexed: 10/10/2024] Open
Abstract
The TGF-β superfamily plays a pivotal role in the regulation of adipogenesis, but little is known about the potential differential role of the three isoforms of TGF-β, TGF-β-1~3. To further elucidate their role, two-dimensionally (2D) and three-dimensionally (3D) cultured 3T3-L1 mouse preadipocytes were subjected to the following analyses: (a) qPCR analysis of adipogenesis-related factors and major extracellular matrix protein (2D and /or 3D), (b) lipid staining by Oil Red O (2D) or BODIPY (3D), (c) Seahorse cellular metabolic measurement (2D), and (d) size and stiffness measurements of 3D 3T3-L1 spheroids. In the 2D cultured 3T3-L1 cells, mRNA expression levels of adipogenesis-related genes and Oil Red O lipid staining intensity were significantly increased by adipogenesis and they were substantially decreased following treatment with 0.1 nm TGF-β isoforms, with TGF-β2 having the greater effects. Consistent with these results, treatment with TGF-β2 resulted in suppression of mitochondrial and glycolytic functions in 2D cultured 3T3-L1 cells. However, the inhibitory effect of TGF-β on adipogenesis decreased under 3D spheroid culture conditions and TGF-β isoforms did not affect adipogenesis-induced (a) enlargement and downsizing of 3T3-L1 spheroids, (b) increase in BODIPY lipid staining intensity, and (c) up-regulation of the mRNA expression of adipogenesis-related genes. The findings presented herein suggest that the three TGF-β isoforms have different suppressive effects on adipogenesis-related cellular properties of 2D cultured 3T3-L1 cells and that their effects decrease under 3D spheroid culture conditions.
Collapse
Affiliation(s)
- Araya Umetsu
- Department of Ophthalmology, Sapporo Medical University School of Medicine, Japan
| | - Megumi Watanabe
- Department of Ophthalmology, Sapporo Medical University School of Medicine, Japan
| | - Tatsuya Sato
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Japan
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, Japan
| | - Megumi Higashide
- Department of Ophthalmology, Sapporo Medical University School of Medicine, Japan
| | - Nami Nishikiori
- Department of Ophthalmology, Sapporo Medical University School of Medicine, Japan
| | - Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Japan
| | - Hiroshi Ohguro
- Department of Ophthalmology, Sapporo Medical University School of Medicine, Japan
| |
Collapse
|
9
|
Okumuş EB, Böke ÖB, Turhan SŞ, Doğan A. From development to future prospects: The adipose tissue & adipose tissue organoids. Life Sci 2024; 351:122758. [PMID: 38823504 DOI: 10.1016/j.lfs.2024.122758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/22/2024] [Accepted: 05/27/2024] [Indexed: 06/03/2024]
Abstract
Living organisms store their energy in different forms of fats including lipid droplets, triacylglycerols, and steryl esters. In mammals and some non-mammal species, the energy is stored in adipose tissue which is the innervated specialized connective tissue that incorporates a variety of cell types such as macrophages, fibroblasts, pericytes, endothelial cells, adipocytes, blood cells, and several kinds of immune cells. Adipose tissue is so complex that the scope of its function is not only limited to energy storage, it also encompasses to thermogenesis, mechanical support, and immune defense. Since defects and complications in adipose tissue are heavily related to certain chronic diseases such as obesity, cardiovascular diseases, type 2 diabetes, insulin resistance, and cholesterol metabolism defects, it is important to further study adipose tissue to enlighten further mechanisms behind those diseases to develop possible therapeutic approaches. Adipose organoids are accepted as very promising tools for studying fat tissue development and its underlying molecular mechanisms, due to their high recapitulation of the adipose tissue in vitro. These organoids can be either derived using stromal vascular fractions or pluripotent stem cells. Due to their great vascularization capacity and previously reported incontrovertible regulatory role in insulin sensitivity and blood glucose levels, adipose organoids hold great potential to become an excellent candidate for the source of stem cell therapy. In this review, adipose tissue types and their corresponding developmental stages and functions, the importance of adipose organoids, and the potential they hold will be discussed in detail.
Collapse
Affiliation(s)
- Ezgi Bulut Okumuş
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, Turkey
| | - Özüm Begüm Böke
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, Turkey
| | - Selinay Şenkal Turhan
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, Turkey
| | - Ayşegül Doğan
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, Turkey.
| |
Collapse
|
10
|
Rapuano R, Mercuri A, Dallavalle S, Moricca S, Lavecchia A, Lupo A. Cladosporols and PPARγ: Same Gun, Same Bullet, More Targets. Biomolecules 2024; 14:998. [PMID: 39199386 PMCID: PMC11353246 DOI: 10.3390/biom14080998] [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: 07/01/2024] [Revised: 07/26/2024] [Accepted: 08/02/2024] [Indexed: 09/01/2024] Open
Abstract
Several natural compounds have been found to act as PPARγ agonists, thus regulating numerous biological processes, including the metabolism of carbohydrates and lipids, cell proliferation and differentiation, angiogenesis, and inflammation. Recently, Cladosporols, secondary metabolites purified from the fungus Cladosporium tenuissimum, have been demonstrated to display an efficient ability to control cell proliferation in human colorectal and prostate cancer cells through a PPARγ-mediated modulation of gene expression. In addition, Cladosporols exhibited a strong anti-adipogenetic activity in 3T3-L1 murine preadipocytes, preventing their in vitro differentiation into mature adipocytes. These data interestingly point out that the interaction between Cladosporols and PPARγ, in the milieu of different cells or tissues, might generate a wide range of beneficial effects for the entire organism affected by diabetes, obesity, inflammation, and cancer. This review explores the molecular mechanisms by which the Cladosporol/PPARγ complex may simultaneously interfere with a dysregulated lipid metabolism and cancer promotion and progression, highlighting the potential therapeutic benefits of Cladosporols for human health.
Collapse
Affiliation(s)
- Roberta Rapuano
- Dipartimento di Scienze e Tecnologie, Università del Sannio, Via dei Mulini, 82100 Benevento, Italy; (R.R.); (A.M.)
| | - Antonella Mercuri
- Dipartimento di Scienze e Tecnologie, Università del Sannio, Via dei Mulini, 82100 Benevento, Italy; (R.R.); (A.M.)
| | - Sabrina Dallavalle
- Dipartimento di Scienze per gli Alimenti, la Nutrizione e l’Ambiente, Università degli Studi di Milano, Via Celoria 2, 20133 Milano, Italy;
| | - Salvatore Moricca
- Dipartimento di Scienze e Tecnologie Agrarie, Alimentari, Ambientali e Forestali (DAGRI), Università degli Studi di Firenze, Piazzale delle Cascine 28, 50144 Firenze, Italy;
| | - Antonio Lavecchia
- Dipartimento di Farmacia “Drug Discovery Laboratory”, Università di Napoli “Federico II”, Via D. Montesano 49, 80131 Napoli, Italy
| | - Angelo Lupo
- Dipartimento di Scienze e Tecnologie, Università del Sannio, Via dei Mulini, 82100 Benevento, Italy; (R.R.); (A.M.)
| |
Collapse
|
11
|
Choi JW, Park GH, Choi HJ, Lee JW, Kwon HY, Choi MY, Jeong JB. Anti‑obesity and immunostimulatory activity of Chrysosplenium flagelliferum in mouse preadipocytes 3T3‑L1 cells and mouse macrophage RAW264.7 cells. Exp Ther Med 2024; 28:315. [PMID: 38911047 PMCID: PMC11190883 DOI: 10.3892/etm.2024.12604] [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: 04/04/2024] [Accepted: 05/22/2024] [Indexed: 06/25/2024] Open
Abstract
Chrysosplenium flagelliferum (CF) is known for its anti-inflammatory, antioxidant and antibacterial activities. However, there is a lack of research on its other pharmacological properties. In the present study, the bifunctional roles of CF in 3T3-L1 and RAW264.7 cells were investigated, focusing on its anti-obesity and immunostimulatory effects. In 3T3-L1 cells, CF effectively mitigated the accumulation of lipid droplets and triacylglycerol. Additionally, CF downregulated the peroxisome proliferator-activated receptor (PPAR)-γ and CCAAT/enhancer-binding protein α protein levels; however, this effect was impeded by the knockdown of β-catenin using β-catenin-specific small interfering RNA. Consequently, CF-mediated inhibition of lipid accumulation was also decreased. CF increased the protein levels of adipose triglyceride lipase and phosphorylated hormone-sensitive lipase, while decreasing those of perilipin-1. Moreover, CF elevated the protein levels of phosphorylated AMP-activated protein kinase and PPARγ coactivator 1-α. In RAW264.7 cells, CF enhanced the production of pro-inflammatory mediators, such as nitric oxide (NO), inducible NO synthase, interleukin (IL)-1β, IL-6 and tumor necrosis factor-α, and increased their phagocytic capacities. Inhibition of Toll-like receptor (TLR)-4 significantly reduced the effects of CF on the production of pro-inflammatory mediators and phagocytosis, indicating its crucial role in facilitating these effects. CF-induced increase in the production of pro-inflammatory mediators was controlled by the activation of c-Jun N-terminal kinase (JNK) and nuclear factor (NF)-κB pathways, and TLR4 inhibition attenuated the phosphorylation of these kinases. The results of the pesent study suggested that CF inhibits lipid accumulation by suppressing adipogenesis and inducing lipolysis and thermogenesis in 3T3-L1 cells, while stimulating macrophage activation via the activation of JNK and NF-κB signaling pathways mediated by TLR4 in RAW264.7 cells. Therefore, CF simultaneously exerts both anti-obesity and immunostimulatory effects.
Collapse
Affiliation(s)
- Jeong Won Choi
- Department of Forest Science, Andong National University, Andong, Gyeongsangbuk 36729, Republic of Korea
| | - Gwang Hun Park
- Forest Medicinal Resources Research Center, National Institute of Forest Science, Yeongju, Gyeongsangbuk 36040, Republic of Korea
| | - Hyeok Jin Choi
- Department of Forest Science, Andong National University, Andong, Gyeongsangbuk 36729, Republic of Korea
| | - Jae Won Lee
- Department of Forest Science, Andong National University, Andong, Gyeongsangbuk 36729, Republic of Korea
| | - Hae-Yun Kwon
- Forest Medicinal Resources Research Center, National Institute of Forest Science, Yeongju, Gyeongsangbuk 36040, Republic of Korea
| | - Min Yeong Choi
- Forest Medicinal Resources Research Center, National Institute of Forest Science, Yeongju, Gyeongsangbuk 36040, Republic of Korea
| | - Jin Boo Jeong
- Department of Forest Science, Andong National University, Andong, Gyeongsangbuk 36729, Republic of Korea
| |
Collapse
|
12
|
Kim HY, Jang HJ, Muthamil S, Shin UC, Lyu JH, Kim SW, Go Y, Park SH, Lee HG, Park JH. Novel insights into regulators and functional modulators of adipogenesis. Biomed Pharmacother 2024; 177:117073. [PMID: 38981239 DOI: 10.1016/j.biopha.2024.117073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/27/2024] [Accepted: 06/29/2024] [Indexed: 07/11/2024] Open
Abstract
Adipogenesis is a process that differentiates new adipocytes from precursor cells and is tightly regulated by several factors, including many transcription factors and various post-translational modifications. Recently, new roles of adipogenesis have been suggested in various diseases. However, the molecular mechanisms and functional modulation of these adipogenic genes remain poorly understood. This review summarizes the regulatory factors and modulators of adipogenesis and discusses future research directions to identify novel mechanisms regulating adipogenesis and the effects of adipogenic regulators in pathological conditions. The master adipogenic transcriptional factors PPARγ and C/EBPα were identified along with other crucial regulatory factors such as SREBP, Kroxs, STAT5, Wnt, FOXO1, SWI/SNF, KLFs, and PARPs. These transcriptional factors regulate adipogenesis through specific mechanisms, depending on the adipogenic stage. However, further studies related to the in vivo role of newly discovered adipogenic regulators and their function in various diseases are needed to develop new potent therapeutic strategies for metabolic diseases and cancer.
Collapse
Affiliation(s)
- Hyun-Yong Kim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea; New Drug Development Center, Osong Medical Innovation Foundation, 123, Osongsaengmyeong-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, Republic of Korea.
| | - Hyun-Jun Jang
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea; Research Group of Personalized Diet, Korea Food Research Institute, Wanju-gun, Jeollabuk-do 55365, Republic of Korea.
| | - Subramanian Muthamil
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea.
| | - Ung Cheol Shin
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea.
| | - Ji-Hyo Lyu
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea.
| | - Seon-Wook Kim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea.
| | - Younghoon Go
- Korean Medicine (KM)-application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea.
| | - Seong-Hoon Park
- Genetic and Epigenetic Toxicology Research Group, Korea Institute of Toxicology, Daejeon 34141, Republic of Korea.
| | - Hee Gu Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea.
| | - Jun Hong Park
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea; University of Science & Technology (UST), KIOM campus, Korean Convergence Medicine Major, Daejeon 34054, Republic of Korea.
| |
Collapse
|
13
|
Prapaharan B, Lea M, Beaudry JL. Weighing in on the role of brown adipose tissue for treatment of obesity. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2024; 27:13157. [PMID: 39087083 PMCID: PMC11290130 DOI: 10.3389/jpps.2024.13157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024]
Abstract
Brown adipose tissue (BAT) activation is an emerging target for obesity treatments due to its thermogenic properties stemming from its ability to shuttle energy through uncoupling protein 1 (Ucp1). Recent rodent studies show how BAT and white adipose tissue (WAT) activity can be modulated to increase the expression of thermogenic proteins. Consequently, these alterations enable organisms to endure cold-temperatures and elevate energy expenditure, thereby promoting weight loss. In humans, BAT is less abundant in obese subjects and impacts of thermogenesis are less pronounced, bringing into question whether energy expending properties of BAT seen in rodents can be translated to human models. Our review will discuss pharmacological, hormonal, bioactive, sex-specific and environmental activators and inhibitors of BAT to determine the potential for BAT to act as a therapeutic strategy. We aim to address the feasibility of utilizing BAT modulators for weight reduction in obese individuals, as recent studies suggest that BAT's contributions to energy expenditure along with Ucp1-dependent and -independent pathways may or may not rectify energy imbalance characteristic of obesity.
Collapse
Affiliation(s)
| | | | - Jacqueline L. Beaudry
- Temerty Faculty of Medicine, Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
14
|
Iacobini C, Vitale M, Haxhi J, Menini S, Pugliese G. Impaired Remodeling of White Adipose Tissue in Obesity and Aging: From Defective Adipogenesis to Adipose Organ Dysfunction. Cells 2024; 13:763. [PMID: 38727299 PMCID: PMC11083890 DOI: 10.3390/cells13090763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
The adipose organ adapts and responds to internal and environmental stimuli by remodeling both its cellular and extracellular components. Under conditions of energy surplus, the subcutaneous white adipose tissue (WAT) is capable of expanding through the enlargement of existing adipocytes (hypertrophy), followed by de novo adipogenesis (hyperplasia), which is impaired in hypertrophic obesity. However, an impaired hyperplastic response may result from various defects in adipogenesis, leading to different WAT features and metabolic consequences, as discussed here by reviewing the results of the studies in animal models with either overexpression or knockdown of the main molecular regulators of the two steps of the adipogenesis process. Moreover, impaired WAT remodeling with aging has been associated with various age-related conditions and reduced lifespan expectancy. Here, we delve into the latest advancements in comprehending the molecular and cellular processes underlying age-related changes in WAT function, their involvement in common aging pathologies, and their potential as therapeutic targets to influence both the health of elderly people and longevity. Overall, this review aims to encourage research on the mechanisms of WAT maladaptation common to conditions of both excessive and insufficient fat tissue. The goal is to devise adipocyte-targeted therapies that are effective against both obesity- and age-related disorders.
Collapse
|
15
|
Moreira LR, Silva AC, da Costa-Oliveira CN, da Silva-Júnior CD, Oliveira KKDS, Torres DJL, Barros MD, Rabello MCDS, de Lorena VMB. Interaction between peripheral blood mononuclear cells and Trypanosoma cruzi-infected adipocytes: implications for treatment failure and induction of immunomodulatory mechanisms in adipose tissue. Front Immunol 2024; 15:1280877. [PMID: 38533504 PMCID: PMC10963431 DOI: 10.3389/fimmu.2024.1280877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 02/27/2024] [Indexed: 03/28/2024] Open
Abstract
Background/Introduction Adipose tissue (AT) has been highlighted as a promising reservoir of infection for viruses, bacteria and parasites. Among them is Trypanosoma cruzi, which causes Chagas disease. The recommended treatment for the disease in Brazil is Benznidazole (BZ). However, its efficacy may vary according to the stage of the disease, geographical origin, age, immune background of the host and sensitivity of the strains to the drug. In this context, AT may act as an ally for the parasite survival and persistence in the host and a barrier for BZ action. Therefore, we investigated the immunomodulation of T. cruzi-infected human AT in the presence of peripheral blood mononuclear cells (PBMC) where BZ treatment was added. Methods We performed indirect cultivation between T. cruzi-infected adipocytes, PBMC and the addition of BZ. After 72h of treatment, the supernatant was collected for cytokine, chemokine and adipokine assay. Infected adipocytes were removed to quantify T. cruzi DNA, and PBMC were removed for immunophenotyping. Results Our findings showed elevated secretion of interleukin (IL)-6, IL-2 and monocyte chemoattractant protein-1 (MCP-1/CCL2) in the AT+PBMC condition compared to the other controls. In contrast, there was a decrease in tumor necrosis factor (TNF) and IL-8/CXCL-8 in the groups with AT. We also found high adipsin secretion in PBMC+AT+T compared to the treated condition (PBMC+AT+T+BZ). Likewise, the expression of CD80+ and HLA-DR+ in CD14+ cells decreased in the presence of T. cruzi. Discussion Thus, our findings indicate that AT promotes up-regulation of inflammatory products such as IL-6, IL-2, and MCP-1/CCL2. However, adipogenic inducers may have triggered the downregulation of TNF and IL-8/CXCL8 through the peroxisome proliferator agonist gamma (PPAR-g) or receptor expression. On the other hand, the administration of BZ only managed to reduce inflammation in the microenvironment by decreasing adipsin in the infected culture conditions. Therefore, given the findings, we can see that AT is an ally of the parasite in evading the host's immune response and the pharmacological action of BZ.
Collapse
Affiliation(s)
- Leyllane Rafael Moreira
- Department of Tropical Medicine, Federal University of Pernambuco, Recife, Brazil
- Department of Immunology, Aggeu Magalhães Institute, Recife, Brazil
| | - Ana Carla Silva
- Department of Immunology, Aggeu Magalhães Institute, Recife, Brazil
| | | | - Claudeir Dias da Silva-Júnior
- Department of Tropical Medicine, Federal University of Pernambuco, Recife, Brazil
- Department of Immunology, Aggeu Magalhães Institute, Recife, Brazil
| | | | - Diego José Lira Torres
- Department of Tropical Medicine, Federal University of Pernambuco, Recife, Brazil
- Department of Immunology, Aggeu Magalhães Institute, Recife, Brazil
| | | | | | | |
Collapse
|
16
|
Xu X, Charrier A, Congrove S, Buchner DA. Cell-state dependent regulation of PPAR γ signaling by ZBTB9 in adipocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.04.583402. [PMID: 38496622 PMCID: PMC10942320 DOI: 10.1101/2024.03.04.583402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Adipocytes play a critical role in metabolic homeostasis. Peroxisome proliferator-activated receptor- γ (PPAR γ ) is a nuclear hormone receptor that is a master regulator of adipocyte differentiation and function. ZBTB9 was predicted to interact with PPAR γ based on large-scale protein interaction experiments. In addition, GWAS studies in the type 2 diabetes (T2D) Knowledge Portal revealed associations between Z btb9 and both BMI and T2D risk. Here we show that ZBTB9 positively regulates PPAR γ activity in mature adipocytes. Surprisingly Z btb9 knockdown (KD) also increased adipogenesis in 3T3-L1 cells and human preadipocytes. E2F activity was increased and E2F downstream target genes were upregulated in Zbtb9 -KD preadipocytes. Accordingly, RB phosphorylation, which regulates E2F activity, was enhanced in Zbtb9 -KD preadipocytes. Critically, an E2F1 inhibitor blocked the effects of Zbtb9 deficiency on adipogenic gene expression and lipid accumulation. Collectively, these results demonstrate that Zbtb9 inhibits adipogenesis as a negative regulator of Pparg expression via altered RB-E2F1 signaling. Our findings reveal complex cell-state dependent roles of ZBTB9 in adipocytes, identifying a new molecule that regulates adipogenesis and adipocyte biology as both a positive and negative regulator of PPAR γ signaling depending on the cellular context, and thus may be important in the pathogenesis and treatment of obesity and T2D.
Collapse
|
17
|
Kuhlmann-Hogan A, Cordes T, Xu Z, Kuna RS, Traina KA, Robles-Oteíza C, Ayeni D, Kwong EM, Levy S, Globig AM, Nobari MM, Cheng GZ, Leibel SL, Homer RJ, Shaw RJ, Metallo CM, Politi K, Kaech SM. EGFR-driven lung adenocarcinomas coopt alveolar macrophage metabolism and function to support EGFR signaling and growth. Cancer Discov 2024; 14:733526. [PMID: 38241033 PMCID: PMC11258210 DOI: 10.1158/2159-8290.cd-23-0434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 11/15/2023] [Accepted: 12/21/2023] [Indexed: 01/26/2024]
Abstract
The limited efficacy of currently approved immunotherapies in EGFR-driven lung adenocarcinoma (LUAD) underscores the need to better understand alternative mechanisms governing local immunosuppression to fuel novel therapies. Elevated surfactant and GM-CSF secretion from the transformed epithelium induces tumor-associated alveolar macrophage (TA-AM) proliferation which supports tumor growth by rewiring inflammatory functions and lipid metabolism. TA-AM properties are driven by increased GM-CSF-PPARγ signaling and inhibition of airway GM-CSF or PPARγ in TA-AMs suppresses cholesterol efflux to tumor cells, which impairs EGFR phosphorylation and restrains LUAD progression. In the absence of TA-AM metabolic support, LUAD cells compensate by increasing cholesterol synthesis, and blocking PPARγ in TA-AMs simultaneous with statin therapy further suppresses tumor progression and increases proinflammatory immune responses. These results reveal new therapeutic combinations for immunotherapy resistant EGFR-mutant LUADs and demonstrate how cancer cells can metabolically co-opt TA-AMs through GM-CSF-PPARγ signaling to provide nutrients that promote oncogenic signaling and growth.
Collapse
Affiliation(s)
- Alexandra Kuhlmann-Hogan
- Department of Immunobiology, Yale School of Medicine, New Haven, CT
- Nomis Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA
| | - Thekla Cordes
- Molecular and Cellular Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA
- Department of Bioinformatics and Biochemistry, Braunshweig Integrated Centre of Systems Biology (BRICS), Technishe Universität Braunschweig, Germany
- Research Group Cellular Metabolism in Infection, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Ziyan Xu
- Nomis Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA
- Division of Biological Sciences, University of California San Diego, La Jolla, CA
| | - Ramya S. Kuna
- Molecular and Cellular Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA
| | - Kacie A. Traina
- Nomis Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA
| | | | - Deborah Ayeni
- Departments of Pathology and Internal Medicine, (Section of Medical Oncology), Yale School of Medicine, New Haven, CT
| | - Elizabeth M. Kwong
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, CA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA
| | - Stellar Levy
- Departments of Pathology and Internal Medicine, (Section of Medical Oncology), Yale School of Medicine, New Haven, CT
| | - Anna-Maria Globig
- Nomis Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA
| | - Matthew M. Nobari
- Division of Pulmonary and Critical Sleep Medicine, University of California San Diego Department of Medicine, La Jolla, CA
| | - George Z. Cheng
- Division of Pulmonary and Critical Sleep Medicine, University of California San Diego Department of Medicine, La Jolla, CA
| | - Sandra L. Leibel
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, CA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA
| | - Robert J. Homer
- Departments of Pathology and Internal Medicine (Section of Pulmonary, Critical Care and Sleep Medicine), Yale University School of Medicine, New Haven, CT
| | - Reuben J. Shaw
- Molecular and Cellular Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA
| | - Christian M. Metallo
- Molecular and Cellular Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA
| | - Katerina Politi
- Departments of Pathology and Internal Medicine, (Section of Medical Oncology), Yale School of Medicine, New Haven, CT
- Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Susan M. Kaech
- Nomis Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA
| |
Collapse
|
18
|
Ma M, Cao R, Tian Y, Fu X. Ubiquitination and Metabolic Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1466:47-79. [PMID: 39546135 DOI: 10.1007/978-981-97-7288-9_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
The increasing incidence of metabolic diseases, including obesity, type 2 diabetes mellitus (T2DM), and non-alcoholic fatty liver disease (NAFLD), in the past decade is a serious concern worldwide. Disruption of cellular protein homeostasis has been considered as a crucial contributor to the pathogenesis of metabolic diseases. To maintain protein homeostasis, cells have evolved multiple dynamic and self-regulating quality control processes to adapt new environmental conditions and prevent prolonged damage. Among them, the ubiquitin proteasome system (UPS), the primary proteolytic pathway for degradation of aberrant proteins via ubiquitination, has an essential role in maintaining cellular homeostasis in response to intracellular stress. Correspondingly, accumulating evidences have shown that dysregulation of ubiquitination can aggravate various metabolic derangements in many tissues, including the liver, skeletal muscle, pancreas, and adipose tissue, and is involved in the initiation and progression of diverse metabolic diseases. In this part, we will summarize the role of ubiquitination in the pathogenesis of metabolic diseases, including obesity, T2DM and NAFLD, and discuss its potential as a therapeutic target.
Collapse
Affiliation(s)
- Meilin Ma
- Division of Endocrinology and Metabolism, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Rong Cao
- Division of Endocrinology and Metabolism, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Yan Tian
- Division of Endocrinology and Metabolism, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Xianghui Fu
- State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
19
|
Celık ZM, Sargin M, Tamer HG, Gunes FE. The effect of lyophilized dried cornelian cherry ( Cornus mas L.) intake on anthropometric and biochemical parameters in women with insulin resistance: A randomized controlled trial. Food Sci Nutr 2023; 11:8060-8071. [PMID: 38107130 PMCID: PMC10724626 DOI: 10.1002/fsn3.3725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 12/19/2023] Open
Abstract
The aim of this study was to determine the effect on the anthropometric and biochemical parameters for women with insulin resistance when lyophilized dried cornelian cherry (Cornus mas L., CM) was added to medical nutrition therapy (MNT). The study was conducted with 84 women aged 18-45, who had been diagnosed with insulin resistance. Participants were randomized into four groups: MNT + 20 g lyophilized dried CM group (DCm, n = 22), MNT group (D, n = 21), only 20 g lyophilized dried CM group (Cm, n = 21), and the control group (C, n = 20). All participants were followed for 12 weeks. While pre- and post-intervention biochemical parameters were recorded from patient files, anthropometric measurements and food consumption records were taken every 15 days. Pre-intervention groups were homogeneously distributed. Post-intervention, among the groups, all anthropometric measurements were similar between the DCm and D, while the percentage of decrease in insulin resistance-related parameters was approximately two times greater in DCm than in D (p < .05). When the Cm and C were compared, it was found that all post-intervention anthropometric measurements were similar, but the percentage of decrease in fasting blood glucose, fasting insulin, and HOMA-IR (Homeostasis Model Assessment-Insulin Resistance) values were greater in C (p < .05). In this study, it was concluded that CM consumption resulted with a decrease in insulin resistance-related biochemical parameters independent of body weight change. Nevertheless, MNT has positive effects on women with insulin resistance, and adding lyophilized dried CM to MNT improves insulin resistance-related parameters and may be beneficial for preventing the development of diabetes.
Collapse
Affiliation(s)
- Zehra Margot Celık
- Department of Nutrition and DieteticsMarmara University Faculty of Health SciencesIstanbulTurkey
| | - Mehmet Sargin
- Faculty of Medicine, Family MedicineIstanbul Medeniyet UniversityIstanbulTurkey
| | - Havva Gonca Tamer
- Faculty of Medicine, Internal MedicineIstanbul Medeniyet UniversityIstanbulTurkey
| | - Fatma Esra Gunes
- Department of Nutrition and Dietetics, Faculty of Health SciencesIstanbul Medeniyet UniversityIstanbulTurkey
| |
Collapse
|
20
|
Zhang J, Zhang Y, Zhou Y, Zhao W, Li J, Yang D, Xiang L, Du T, Ma L. Effect of vitamin D3 on lipid droplet growth in adipocytes of mice with HFD-induced obesity. Food Sci Nutr 2023; 11:6686-6697. [PMID: 37823117 PMCID: PMC10563741 DOI: 10.1002/fsn3.3618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 07/16/2023] [Accepted: 07/29/2023] [Indexed: 10/13/2023] Open
Abstract
Vitamin D-regulating action of PPARγ on obesity has been confirmed on adipocyte differentiation. However, it is not clear whether vitamin D affects the morphological size of mature adipocytes by influencing the expression of PPARγ in vivo. Our hypothesis was that Vitamin D3 (VitD3) inhibits the growth of adipocyte size by suppressing PPARγ expression in white adipocytes of obese mice. Five-week-old male C57BL/6J mice were randomly divided into normal diet and high-fat diet groups. After 10 weeks, the body weight between the two groups differed by 26.91%. The obese mice were randomly divided into a high-fat diet, solvent control, low-dose VitD3 (5000 IU/kg·food), medium-dose VitD3 (7500 IU/kg·food), high-dose VitD3 (10,000 IU/kg·food), and PPAR γ antagonist group, and the intervention lasted for 8 weeks. Diet-induced obesity (DIO) mice fed high-dose VitD3 exacerbated markers of adiposity (body weight, fat mass, fat mass rate, size of white and brown adipocytes, mRNA, and protein levels of ATGL and Fsp27), and the protein level of ATGL and Fsp27 decreased in the low-dose group. In conclusion, high-dose VitD3 possibly via inhibiting the ATGL expression, thereby inhibiting lipolysis, increasing the volume of adipocytes, and decreasing their fat-storing ability resulted in decreased Fsp27 expression. Therefore, long-term high-dose oral VitD3 may not necessarily improve obesity, and we need more clinical trials to explore the intervention dose and duration of VitD3 in the treatment of VitD3 deficiency in obese patients.
Collapse
Affiliation(s)
- Jingjing Zhang
- Department of Clinical NutritionAffiliated Hospital of Southwest Medical UniversityLuzhouSichuanChina
| | - Yuanfan Zhang
- Department of Nutrition and Food Hygiene, School of Public HealthSouthwest Medical UniversityLuzhouSichuanChina
| | - Yong Zhou
- Department of Medical Cell Biology and Genetics, School of Basic Medical ScienceSouthwest Medical UniversityLuzhouSichuanChina
| | - Wenxin Zhao
- Department of Nutrition and Food Hygiene, School of Public HealthSouthwest Medical UniversityLuzhouSichuanChina
| | - Jialu Li
- Department of Nutrition and Food Hygiene, School of Public HealthSouthwest Medical UniversityLuzhouSichuanChina
| | - Dan Yang
- Department of Nutrition and Food Hygiene, School of Public HealthSouthwest Medical UniversityLuzhouSichuanChina
| | - Lian Xiang
- Department of Nutrition and Food Hygiene, School of Public HealthSouthwest Medical UniversityLuzhouSichuanChina
| | - Tingwan Du
- Department of Nutrition and Food Hygiene, School of Public HealthSouthwest Medical UniversityLuzhouSichuanChina
| | - Ling Ma
- Department of Nutrition and Food Hygiene, School of Public HealthSouthwest Medical UniversityLuzhouSichuanChina
| |
Collapse
|
21
|
Jeong JH, Lee HL, Park HJ, Yoon YE, Shin J, Jeong MY, Park SH, Kim DH, Han SW, Kang CG, Hong KJ, Lee SJ. Effects of tomato ketchup and tomato paste extract on hepatic lipid accumulation and adipogenesis. Food Sci Biotechnol 2023; 32:1111-1122. [PMID: 37215254 PMCID: PMC10195947 DOI: 10.1007/s10068-023-01244-x] [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: 09/08/2022] [Revised: 12/09/2022] [Accepted: 01/02/2023] [Indexed: 02/09/2023] Open
Abstract
Tomatoes include high levels of lycopene, which is a potent antioxidative, hypolipidemic, and antidiabetic phytochemical. The intake of lycopene is associated with a reduced risk of insulin resistance and metabolic syndrome. The aim of this study was to investigate whether tomato ketchup and tomato paste, major dietary sources for tomato and lycopene, could regulate hepatic lipid metabolism and adipogenesis. To investigate the regulatory effects of tomato ketchup and tomato paste, we prepared a tomato ketchup extract (TKE) and a tomato paste extract (TPE) in 80% (v/v) ethyl acetate for the experiment. TKE and TPE reduced lipid accumulation and key markers for gluconeogenesis and induced a higher rate of fatty acid oxidation in HepG2 hepatocytes. In 3T3-L1 adipocytes, TKE and TPE increased adipogenesis and intracellular triglyceride accumulation, and stimulated glucose uptake. Peroxisome proliferator-activated receptor alpha and gamma expression levels were increased by TKE and TPE treatment. A single oral dose of tomato ketchup and tomato paste (9.28 g/kg) significantly improved glucose and insulin tolerance in mice. These findings suggest that lycopene-containing tomato ketchup and tomato paste may have beneficial regulatory effects in terms of energy metabolism in hepatocytes and adipocytes, and thus may improve blood glucose metabolism.
Collapse
Affiliation(s)
- Ji Hyun Jeong
- Department of Biotechnology, Graduate School of Life Sciences & Biotechnology, College of Life Sciences & Biotechnology, Korea University, Seoul, 02841 Republic of Korea
| | - Ha Lim Lee
- Department of Biotechnology, Graduate School of Life Sciences & Biotechnology, College of Life Sciences & Biotechnology, Korea University, Seoul, 02841 Republic of Korea
| | - Hyun Ji Park
- Department of Biotechnology, Graduate School of Life Sciences & Biotechnology, College of Life Sciences & Biotechnology, Korea University, Seoul, 02841 Republic of Korea
| | - Ye Eun Yoon
- Department of Biotechnology, Graduate School of Life Sciences & Biotechnology, College of Life Sciences & Biotechnology, Korea University, Seoul, 02841 Republic of Korea
| | - Jaeeun Shin
- Department of Biotechnology, Graduate School of Life Sciences & Biotechnology, College of Life Sciences & Biotechnology, Korea University, Seoul, 02841 Republic of Korea
| | - Mi-Young Jeong
- Department of Biotechnology, Graduate School of Life Sciences & Biotechnology, College of Life Sciences & Biotechnology, Korea University, Seoul, 02841 Republic of Korea
| | - Sung Hoon Park
- Department of Food & Nutrition, College of Life Sciences, Gangneung-Wonju National University, Gangneung, South Korea
| | - Da-hye Kim
- R&D Center, Ottogi Corporation, Anyang-Si, 14060 Republic of Korea
| | - Seung-Woo Han
- R&D Center, Ottogi Corporation, Anyang-Si, 14060 Republic of Korea
| | - Choon-Gil Kang
- R&D Center, Ottogi Corporation, Anyang-Si, 14060 Republic of Korea
| | - Ki-Ju Hong
- R&D Center, Ottogi Corporation, Anyang-Si, 14060 Republic of Korea
| | - Sung-Joon Lee
- Department of Biotechnology, Graduate School of Life Sciences & Biotechnology, College of Life Sciences & Biotechnology, Korea University, Seoul, 02841 Republic of Korea
- Department of Food Bioscience & Technology, College of Life Sciences & Biotechnology, Korea University, Seoul, South Korea
- Interdisciplinary Program in Precision Public Health, BK21 Four Institute of Precision Public Health, Korea University, Seoul, South Korea
| |
Collapse
|
22
|
Kuhlmann-Hogan A, Cordes T, Xu Z, Traina KA, Robles-Oteíza C, Ayeni D, Kwong EM, Levy SR, Nobari M, Cheng GZ, Shaw R, Leibel SL, Metallo CM, Politi K, Kaech SM. EGFR + lung adenocarcinomas coopt alveolar macrophage metabolism and function to support EGFR signaling and growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.15.536974. [PMID: 37131637 PMCID: PMC10153136 DOI: 10.1101/2023.04.15.536974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The limited efficacy of currently approved immunotherapies in EGFR-mutant lung adenocarcinoma (LUAD) underscores the need to better understand mechanisms governing local immunosuppression. Elevated surfactant and GM-CSF secretion from the transformed epithelium induces tumor-associated alveolar macrophages (TA-AM) to proliferate and support tumor growth by rewiring inflammatory functions and lipid metabolism. TA-AM properties are driven by increased GM-CSF-PPARγ signaling and inhibition of airway GM-CSF or PPARγ in TA-AMs suppresses cholesterol efflux to tumor cells, which impairs EGFR phosphorylation and restrains LUAD progression. In the absence of TA-AM metabolic support, LUAD cells compensate by increasing cholesterol synthesis, and blocking PPARγ in TA-AMs simultaneous with statin therapy further suppresses tumor progression and increases T cell effector functions. These results reveal new therapeutic combinations for immunotherapy resistant EGFR-mutant LUADs and demonstrate how such cancer cells can metabolically co-opt TA-AMs through GM-CSF-PPARγ signaling to provide nutrients that promote oncogenic signaling and growth.
Collapse
|
23
|
Xue S, Lee D, Berry DC. Thermogenic adipose tissue in energy regulation and metabolic health. Front Endocrinol (Lausanne) 2023; 14:1150059. [PMID: 37020585 PMCID: PMC10067564 DOI: 10.3389/fendo.2023.1150059] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 03/07/2023] [Indexed: 04/07/2023] Open
Abstract
The ability to generate thermogenic fat could be a targeted therapy to thwart obesity and improve metabolic health. Brown and beige adipocytes are two types of thermogenic fat cells that regulate energy balance. Both adipocytes share common morphological, biochemical, and thermogenic properties. Yet, recent evidence suggests unique features exist between brown and beige adipocytes, such as their cellular origin and thermogenic regulatory processes. Beige adipocytes also appear highly plastic, responding to environmental stimuli and interconverting between beige and white adipocyte states. Additionally, beige adipocytes appear to be metabolically heterogenic and have substrate specificity. Nevertheless, obese and aged individuals cannot develop beige adipocytes in response to thermogenic fat-inducers, creating a key clinical hurdle to their therapeutic promise. Thus, elucidating the underlying developmental, molecular, and functional mechanisms that govern thermogenic fat cells will improve our understanding of systemic energy regulation and strive for new targeted therapies to generate thermogenic fat. This review will examine the recent advances in thermogenic fat biogenesis, molecular regulation, and the potential mechanisms for their failure.
Collapse
Affiliation(s)
| | | | - Daniel C. Berry
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| |
Collapse
|
24
|
Chaves-Filho AB, Peixoto AS, Castro É, Oliveira TE, Perandini LA, Moreira RJ, da Silva RP, da Silva BP, Moretti EH, Steiner AA, Miyamoto S, Yoshinaga MY, Festuccia WT. Futile cycle of β-oxidation and de novo lipogenesis are associated with essential fatty acids depletion in lipoatrophy. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159264. [PMID: 36535597 DOI: 10.1016/j.bbalip.2022.159264] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 11/08/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022]
Abstract
Total absence of adipose tissue (lipoatrophy) is associated with the development of severe metabolic disorders including hepatomegaly and fatty liver. Here, we sought to investigate the impact of severe lipoatrophy induced by deletion of peroxisome proliferator-activated receptor gamma (PPARγ) exclusively in adipocytes on lipid metabolism in mice. Untargeted lipidomics of plasma, gastrocnemius and liver uncovered a systemic depletion of the essential linoleic (LA) and α-linolenic (ALA) fatty acids from several lipid classes (storage lipids, glycerophospholipids, free fatty acids) in lipoatrophic mice. Our data revealed that such essential fatty acid depletion was linked to increased: 1) capacity for liver mitochondrial fatty acid β-oxidation (FAO), 2) citrate synthase activity and coenzyme Q content in the liver, 3) whole-body oxygen consumption and reduced respiratory exchange rate in the dark period, and 4) de novo lipogenesis and carbon flux in the TCA cycle. The key role of de novo lipogenesis in hepatic steatosis was evidenced by an accumulation of stearic, oleic, sapienic and mead acids in liver. Our results thus indicate that the simultaneous activation of the antagonic processes FAO and de novo lipogenesis in liver may create a futile metabolic cycle leading to a preferential depletion of LA and ALA. Noteworthy, this previously unrecognized cycle may also explain the increased energy expenditure displayed by lipoatrophic mice, adding a new piece to the metabolic regulation puzzle in lipoatrophies.
Collapse
Affiliation(s)
- Adriano B Chaves-Filho
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Av. Prof Lineu Prestes 748, São Paulo 05508900, Brazil; Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof Lineu Prestes 1524, São Paulo 05508000, Brazil.
| | - Albert S Peixoto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Av. Prof Lineu Prestes 748, São Paulo 05508900, Brazil
| | - Érique Castro
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Av. Prof Lineu Prestes 748, São Paulo 05508900, Brazil
| | - Tiago E Oliveira
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Av. Prof Lineu Prestes 748, São Paulo 05508900, Brazil
| | - Luiz A Perandini
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Av. Prof Lineu Prestes 748, São Paulo 05508900, Brazil
| | - Rafael J Moreira
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Av. Prof Lineu Prestes 748, São Paulo 05508900, Brazil
| | - Railmara P da Silva
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof Lineu Prestes 1524, São Paulo 05508000, Brazil
| | - Beatriz P da Silva
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof Lineu Prestes 1524, São Paulo 05508000, Brazil
| | - Eduardo H Moretti
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, Av. Prof Lineu Prestes 1524, São Paulo 05508000, Brazil
| | - Alexandre A Steiner
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, Av. Prof Lineu Prestes 1524, São Paulo 05508000, Brazil
| | - Sayuri Miyamoto
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof Lineu Prestes 1524, São Paulo 05508000, Brazil
| | - Marcos Y Yoshinaga
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof Lineu Prestes 1524, São Paulo 05508000, Brazil.
| | - William T Festuccia
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Av. Prof Lineu Prestes 748, São Paulo 05508900, Brazil.
| |
Collapse
|
25
|
The Potential Roles of Post-Translational Modifications of PPARγ in Treating Diabetes. Biomolecules 2022; 12:biom12121832. [PMID: 36551260 PMCID: PMC9775095 DOI: 10.3390/biom12121832] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/04/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
The number of patients with type 2 diabetes mellitus (T2DM), which is mainly characterized by insulin resistance and insulin secretion deficiency, has been soaring in recent years. Accompanied by many other metabolic syndromes, such as cardiovascular diseases, T2DM represents a big challenge to public health and economic development. Peroxisome proliferator-activated receptor γ (PPARγ), a ligand-activated nuclear receptor that is critical in regulating glucose and lipid metabolism, has been developed as a powerful drug target for T2DM, such as thiazolidinediones (TZDs). Despite thiazolidinediones (TZDs), a class of PPARγ agonists, having been proven to be potent insulin sensitizers, their use is restricted in the treatment of diabetes for their adverse effects. Post-translational modifications (PTMs) have shed light on the selective activation of PPARγ, which shows great potential to circumvent TZDs' side effects while maintaining insulin sensitization. In this review, we will focus on the potential effects of PTMs of PPARγ on treating T2DM in terms of phosphorylation, acetylation, ubiquitination, SUMOylation, O-GlcNAcylation, and S-nitrosylation. A better understanding of PTMs of PPARγ will help to design a new generation of safer compounds targeting PPARγ to treat type 2 diabetes.
Collapse
|
26
|
Li Z, Bagchi DP, Zhu J, Bowers E, Yu H, Hardij J, Mori H, Granger K, Skjaerlund J, Mandair G, Abrishami S, Singer K, Hankenson KD, Rosen CJ, MacDougald OA. Constitutive bone marrow adipocytes suppress local bone formation. JCI Insight 2022; 7:160915. [PMID: 36048537 PMCID: PMC9675472 DOI: 10.1172/jci.insight.160915] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 08/31/2022] [Indexed: 12/15/2022] Open
Abstract
BM adipocytes (BMAd) are a unique cell population derived from BM mesenchymal progenitors and marrow adipogenic lineage precursors. Although they have long been considered to be a space filler within bone cavities, recent studies have revealed important physiological roles in hematopoiesis and bone metabolism. To date, the approaches used to study BMAd function have been confounded by contributions by nonmarrow adipocytes or by BM stromal cells. To address this gap in the field, we have developed a BMAd-specific Cre mouse model to deplete BMAds by expression of diphtheria toxin A (DTA) or by deletion of peroxisome proliferator-activated receptor gamma (Pparg). We found that DTA-induced loss of BMAds results in decreased hematopoietic stem and progenitor cell numbers and increased bone mass in BMAd-enriched locations, including the distal tibiae and caudal vertebrae. Elevated bone mass appears to be secondary to enhanced endosteal bone formation, suggesting a local effect caused by depletion of BMAd. Augmented bone formation with BMAd depletion protects mice from bone loss induced by caloric restriction or ovariectomy, and it facilitates the bone-healing process after fracture. Finally, ablation of Pparg also reduces BMAd numbers and largely recapitulates high-bone mass phenotypes observed with DTA-induced BMAd depletion.
Collapse
Affiliation(s)
- Ziru Li
- Department of Molecular & Integrative Physiology and
| | | | - Junxiong Zhu
- Department of Orthopedic Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Emily Bowers
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Hui Yu
- Department of Molecular & Integrative Physiology and
| | - Julie Hardij
- Department of Molecular & Integrative Physiology and
| | - Hiroyuki Mori
- Department of Molecular & Integrative Physiology and
| | | | - Jon Skjaerlund
- Department of Orthopedic Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Gurjit Mandair
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Simin Abrishami
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Kanakadurga Singer
- Department of Molecular & Integrative Physiology and
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Kurt D. Hankenson
- Department of Orthopedic Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | - Ormond A. MacDougald
- Department of Molecular & Integrative Physiology and
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
27
|
Chen X, Liu L, Zeng Y, Li D, Liu X, Hu C. Olanzapine induces weight gain in offspring of prenatally exposed poly I:C rats by reducing brown fat thermogenic activity. Front Pharmacol 2022; 13:1001919. [PMID: 36249777 PMCID: PMC9561095 DOI: 10.3389/fphar.2022.1001919] [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: 07/24/2022] [Accepted: 09/13/2022] [Indexed: 11/25/2022] Open
Abstract
Background: Olanzapine (OLZ) is an antipsychotic with a high risk of metabolic syndrome, and its induced metabolic disturbance may be related to the thermogenic function of brown adipose tissue (BAT). Of note is that schizophrenia itself appears to be associated with a higher incidence of metabolic syndrome. However, whether OLZ affects metabolic disorders by regulating BAT function and its mechanism in animal models of schizophrenia have not been reported. Methods: We induced maternal immune activation (MIA) in pregnant rodents by injection of synthetic double-stranded RNA-poly I:C (a virus-like substance), and rats were injected with poly I:C, 10 mg/kg) or saline on day 13 of gestation. Rat offspring received OLZ (1 mg/kg, tid) or vehicle from adulthood for 28 days, and body weight and food intake were recorded. Morphological alterations of white adipose tissue (WAT) and BAT were analyzed by HE and oil red staining, and expression of BAT-specific marker proteins/genes was detected by western blot and qRT-PCR. In addition, embryonic stem cells C3H10T1/2 were used to direct differentiation into brown-like adipocytes, and C3H10T1/2 cells were treated with OLZ for the differentiation process. The effects of OLZ on brown-like adipocyte differentiation and activity were analyzed using oil red staining, immunofluorescence and flow cytometry. Results: Compared with the Veh (saline) group, the TG, pWAT weight, adipocyte size and liver weight of the Veh (poly I:C) group were significantly increased, suggesting that the offspring of Poly I:C rats had obvious dyslipidemia and lipid accumulation, which were risk factors for metabolic abnormalities such as obesity. In addition, OLZ treatment resulted in altered WAT and BAT morphology in poly I:C or saline exposed offspring, causing lipid accumulation and weight gain and reducing the expression of the BAT-specific marker molecule UCP1 protein/gene. At the same time, OLZ inhibited the directional differentiation and mitochondrial activity of C3H10T1/2 brown-like adipocytes. Conclusion: Poly I:C-elicited MIA and OLZ differentially inhibited BAT activity and mitochondrial biogenesis, leading to weight gain in adult rats, a process involving PPAR-γ/UCP1-related thermogenic proteins.
Collapse
Affiliation(s)
- Xiaoying Chen
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
- The Second Affiliated Hospital, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Lu Liu
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
- Department of Preclinical Medicine, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yanping Zeng
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Dejuan Li
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Xuemei Liu
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Changhua Hu
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
- *Correspondence: Changhua Hu,
| |
Collapse
|
28
|
Wang D, Zhang X, Li Y, Jia L, Zhai L, Wei W, Zhang L, Jiang H, Bai Y. Exercise-Induced Browning of White Adipose Tissue and Improving Skeletal Muscle Insulin Sensitivity in Obese/Non-obese Growing Mice: Do Not Neglect Exosomal miR-27a. Front Nutr 2022; 9:940673. [PMID: 35782940 PMCID: PMC9248804 DOI: 10.3389/fnut.2022.940673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Exercise is considered as a favorable measure to prevent and treat childhood obesity. However, the underlying mechanisms of exercise-induced beneficial effects and the difference between obese and non-obese individuals are largely unclear. Recently, miR-27a is recognized as a central upstream regulator of proliferator-activated receptor γ (PPAR-γ) in contributing to various physiological and pathological processes. This study aims to explore the possible cause of exercise affecting white adipose tissue (WAT) browning and reversing skeletal muscle insulin resistance in obese/non-obese immature bodies. For simulating the process of childhood obesity, juvenile mice were fed with a basal diet or high-fat diet (HFD) and took 1 or 2 h swimming exercise simultaneously for 10 weeks. The obese animal model was induced by the HFD. We found that exercise hindered HFD-induced body fat development in growing mice. Exercise modified glucolipid metabolism parameters differently in the obese/non-obese groups, and the changes of the 2 h exercise mice were not consistent with the 1 h exercise mice. The level of serum exosomal miR-27a in the non-exercise obese group was increased obviously, which was reduced in the exercise obese groups. Results from bioinformatics analysis and dual-luciferase reporter assay showed that miR-27a targeted PPAR-γ. Exercise stimulated WAT browning; however, the response of obese WAT lagged behind normal WAT. In the HFD-fed mice, 2 h exercise activated the IRS-1/Akt/GLUT-4 signaling pathway in the skeletal muscles. In summary, our findings confirmed that exercise-induced beneficial effects are associated with exercise duration, and the response of obese and non-obese bodies is different. Exosomal miR-27a might be a crucial node for the process of exercise-induced browning of WAT and improving skeletal muscle insulin sensitivity.
Collapse
Affiliation(s)
- Dongxue Wang
- Department of Maternal and Child Health, School of Public Health, China Medical University, Shenyang, China
- The Second People’s Hospital of Jiashan, Jiaxing, China
| | - Xihuan Zhang
- Department of Maternal and Child Health, School of Public Health, China Medical University, Shenyang, China
- Xinzhou District Center for Disease Control and Prevention, Wuhan, China
| | - Yibai Li
- The First Division of Clinical Medicine, China Medical University, Shenyang, China
| | - Lihong Jia
- Department of Maternal and Child Health, School of Public Health, China Medical University, Shenyang, China
| | - Lingling Zhai
- Department of Maternal and Child Health, School of Public Health, China Medical University, Shenyang, China
| | - Wei Wei
- Department of Maternal and Child Health, School of Public Health, China Medical University, Shenyang, China
| | - Li Zhang
- Department of Dermatology, First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Immunodermatology, Ministry of Education and NHC, National Joint Engineering Research Center for Theranostics of Immunological Skin Diseases, Shenyang, China
| | - Hongkun Jiang
- Department of Pediatrics, First Hospital of China Medical University, Shenyang, China
| | - Yinglong Bai
- Department of Maternal and Child Health, School of Public Health, China Medical University, Shenyang, China
- *Correspondence: Yinglong Bai,
| |
Collapse
|
29
|
Abaj F, Rafiee M, Koohdani F. A Personalized Diet Approach Study: Interaction between PPAR-γ Pro12Ala and Dietary Insulin Indices on Metabolic Markers in Diabetic Patients. J Hum Nutr Diet 2022; 35:663-674. [PMID: 35560467 DOI: 10.1111/jhn.13033] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/05/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND The objectives were to investigate the effect of the interaction between peroxisome proliferator-activated receptor gamma (PPAR-γ) Pro12Ala polymorphisms and dietary insulin load and insulin index (DIL and DII) on Cardio-metabolic Markers among diabetic patients. METHODS This cross-sectional study was conducted on 393 diabetic patients. Food-frequency questionnaire (FFQ) was used for DIL and DII calculation. PPAR-γ Pro12Ala was genotyped by the PCR-RFLP method. Biochemical markers including TC, LDL, HDL, TG, SOD, CRP, TAC, PTX3, PGF2α. IL18, leptin and ghrelin were measured by standard protocol. RESULT Risk-allele carriers (CG, GG) had higher obesity indices WC (P interaction =0.04), BMI (P interaction =0.006) and, WC (P interaction =0.04) compared with individuals with the CC genotype when they consumed a diet with higher DIL and DII respectively. Besides, carriers of the G allele who were in the highest tertile of DIL, had lower HDL (P interaction =0.04) and higher PGF2α (P interaction =0.03) and PTX3 (P interaction =0.03). Moreover, the highest tertile of the DII, showed an increase in IL18 (P interaction =0.01) and lower SOD (P interaction =0.03) for risk allele carriers compared to those with CC homozygotes. CONCLUSION We revealed PPAR-γ Pro12Ala polymorphism was able to intensify the effect of DIL and DII on CVD risk factors; risk-allele carriers who consumed a diet with high DIL and DII score have more likely to be obese and have higher inflammatory markers. Also, protective factor against CVD risk factors were reduced significantly in this group compared to CC homozygotes. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Faezeh Abaj
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Masoumeh Rafiee
- Department of Clinical Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences (IUMS), Isfahan, Iran
| | - Fariba Koohdani
- Department of Cellular, Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
30
|
Brener A, Zeitlin L, Wilnai Y, Birk OS, Rosenfeld T, Chorna E, Lebenthal Y. Looking for the skeleton in the closet-rare genetic diagnoses in patients with diabetes and skeletal manifestations. Acta Diabetol 2022; 59:711-719. [PMID: 35137278 DOI: 10.1007/s00592-022-01854-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/20/2022] [Indexed: 12/12/2022]
Abstract
AIMS The precision medicine approach of tailoring treatment to the individual characteristics of each patient has been a great success in monogenic diabetes subtypes, highlighting the importance of accurate clinical and genetic diagnoses of the type of diabetes. We sought to describe three unique cases of childhood-onset diabetes in whom skeletal manifestations led to the revelation of a rare type of diabetes. METHODS : Case-scenarios and review of the literature. RESULTS Case 1: A homozygous mutation in TRMT10A, a tRNA methyltransferase, was identified in a 15-year-old boy with new-onset diabetes, developmental delay, microcephaly, dysmorphism, short stature and central obesity. The progressive apoptosis of pancreatic beta cells required insulin replacement therapy, with increased demand due to an unfavorable body composition. Case 2: Congenital generalized lipodystrophy type 1 was suspected in an adolescent male with an acromegaloid facial appearance, muscular habitus, and diabetes who presented with a pathological fracture in a cystic bone lesion. A homozygous mutation in AGPAT2, an acyl transferase which mediates the formation of phospholipid precursors, was identified. Leptin replacement therapy initiation resulted in a remarkable improvement in clinical parameters. Case 3: A 12-year-old boy with progressive lower limb weakness and pain was diagnosed with diabetic ketoacidosis. Diffuse diaphyseal osteosclerosis compatible with the diagnosis of Camurati-Engelmann disease and a heterozygous mutation in TGFβ1 were identified. Preservation of euglycemia by insulin replacement relieved pain, suggesting that the diabetic milieu may have augmented TGFβ1 overexpression. CONCLUSION Unraveling the precise genetic cause for the clinical manifestations led to the prediction of phenotypic manifestations, and enhanced the clinical outcomes.
Collapse
Affiliation(s)
- Avivit Brener
- Pediatric Endocrinology and Diabetes Unit, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, 6 Weizmann Street, 6423906, Tel Aviv, Israel.
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Leonid Zeitlin
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Pediatric Orthopedic Department, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Yael Wilnai
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- The Genetics Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Ohad S Birk
- Genetics Institute at Soroka Medical Center and the Morris Kahn Laboratory of Human GeneticsFaculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Talya Rosenfeld
- Pediatric Endocrinology and Diabetes Unit, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, 6 Weizmann Street, 6423906, Tel Aviv, Israel
- The Nutrition & Dietetics Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Efrat Chorna
- Pediatric Endocrinology and Diabetes Unit, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, 6 Weizmann Street, 6423906, Tel Aviv, Israel
- Social Services, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Yael Lebenthal
- Pediatric Endocrinology and Diabetes Unit, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, 6 Weizmann Street, 6423906, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
31
|
Chiang CH, Cheng CY, Lien YT, Huang KC, Lin WW. P2X7 Activation Enhances Lipid Accumulation During Adipocytes Differentiation Through Suppressing the Expression of Sirtuin-3, Sirtuin-5, and Browning Genes. Front Pharmacol 2022; 13:852858. [PMID: 35462937 PMCID: PMC9019299 DOI: 10.3389/fphar.2022.852858] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/18/2022] [Indexed: 11/24/2022] Open
Abstract
P2X7 signaling has been explored in adipose tissue because of its potential to promote ATP-activated inflammatory cascades during obesogenic environments. However, limited literature has investigated the role of the P2X7 receptor in lipid metabolism during adipocyte differentiation. This study sought to explore the regulatory roles of P2X7 in adipocytes. This study utilized the in vitro 3T3-L1 differentiation model. Lipid accumulation, intracellular triglyceride, and extracellular glycerol were determined. The selective P2X7 agonist BzATP and antagonist A438079 were administered to investigate the functions of P2X7. We found that the expression of P2X7 and the lipid accumulation increased during adipocyte differentiation from D0 to D4. When administered at D0/D2, A438079 attenuated, while BzATP enhanced the degree of lipid accumulation during adipocyte differentiation. Neither did BzATP and A438079 administration affect the expression of PPARγ and C/EBPα genes that increased at D4. In addition, both intracellular triglyceride and extracellular glycerol levels at D4 were reduced by A438079 treatment and enhanced by BzATP administration. When administered at stage 2 of adipocyte differentiation, BzATP consistently enhanced lipid accumulation and intracellular triglyceride and extracellular glycerol levels without affecting mRNA and protein levels of PPARγ and C/EBPα that increased at D4. However, treating A438079 or BzATP at D4 did not affect intracellular triglyceride formation and extracellular glycerol release in differentiated adipocytes at D7. Notably, BzATP administration at stage 2 exerted a concentration-dependent inhibition on the enhanced expression of PRDM16, PGC-1α, and UCP-1 at D4. Furthermore, BzATP administration at D0/D2 inhibited the protein and mRNA levels of sirtuin-3/5 at D4. BzATP treatment at stage 2 also suppressed the mRNA levels of sirtuin-3/5 genes upregulated by insulin. In conclusion, this study demonstrated P2X7 enhances lipid accumulation during adipogenesis by suppressing the expression of sirtuin-3/5 and the browning genes.
Collapse
Affiliation(s)
- Chien-Hsieh Chiang
- Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Family Medicine, National Taiwan University Hospital & College of Medicine, Taipei, Taiwan
| | - Ching-Yuan Cheng
- Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yi-Ting Lien
- Department of Family Medicine, National Taiwan University Hospital & College of Medicine, Taipei, Taiwan
| | - Kuo-Chin Huang
- Department of Family Medicine, National Taiwan University Hospital & College of Medicine, Taipei, Taiwan
| | - Wan-Wan Lin
- Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan.,Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
32
|
Zhang Y, Ling L, Ajay D/O Ajayakumar A, Eio YM, van Wijnen AJ, Nurcombe V, Cool SM. FGFR2 accommodates osteogenic cell fate determination in human mesenchymal stem cells. Gene 2022; 818:146199. [PMID: 35093449 PMCID: PMC9256080 DOI: 10.1016/j.gene.2022.146199] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 12/09/2021] [Accepted: 01/13/2022] [Indexed: 01/14/2023]
Abstract
The multilineage differentiation potential of human mesenchymal stem cells (hMSCs) underpins their clinical utility for tissue regeneration. Control of such cell-fate decisions is tightly regulated by different growth factors/cytokines and their cognate receptors. Fibroblast growth factors (FGFs) are among such factors critical for osteogenesis. However, how FGF receptors (FGFRs) help to orchestrate osteogenic progression remains to be fully elucidated. Here, we studied the protein levels of FGFRs during osteogenesis in human adult bone marrow-derived MSCs and discovered a positive correlation between FGFR2 expression and alkaline phosphatase (ALP) activity, an early marker of osteogenesis. Through RNA interference studies, we confirmed the role of FGFR2 in promoting the osteogenic differentiation of hMSCs. Knockdown of FGFR2 resulted in downregulation of pro-osteogenic genes and upregulation of pro-adipogenic genes and adipogenic commitment. Moreover, under osteogenic induction, FGFR2 knockdown resulted in upregulation of Enhancer of Zeste Homolog 2 (EZH2), an epigenetic enzyme that regulates MSC lineage commitment and suppresses osteogenesis. Lastly, we show that serial-passaged hMSCs have reduced FGFR2 expression and impaired osteogenic potential. Our study suggests that FGFR2 is critical for mediating osteogenic fate by regulating the balance of osteo-adipogenic lineage commitment. Therefore, examining FGFR2 levels during serial-passaging of hMSCs may prove useful for monitoring their multipotency.
Collapse
Affiliation(s)
- Ying Zhang
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 138673, Singapore
| | - Ling Ling
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), 138648, Singapore
| | - Arya Ajay D/O Ajayakumar
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 138673, Singapore
| | - Yating Michelle Eio
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 138673, Singapore
| | - Andre J van Wijnen
- Department of Biochemistry, University of Vermont, Burlington, VT 05405, USA
| | - Victor Nurcombe
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), 138648, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University-Imperial College London, 636921, Singapore
| | - Simon M Cool
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 138673, Singapore; Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119288, Singapore.
| |
Collapse
|
33
|
Chen KY, De Angulo A, Guo X, More A, Ochsner SA, Lopez E, Saul D, Pang W, Sun Y, McKenna NJ, Tong Q. Adipocyte-Specific Ablation of PU.1 Promotes Energy Expenditure and Ameliorates Metabolic Syndrome in Aging Mice. FRONTIERS IN AGING 2022; 2:803482. [PMID: 35822007 PMCID: PMC9261351 DOI: 10.3389/fragi.2021.803482] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/13/2021] [Indexed: 12/03/2022]
Abstract
Objective: Although PU.1/Spi1 is known as a master regulator for macrophage development and function, we have reported previously that it is also expressed in adipocytes and is transcriptionally induced in obesity. Here, we investigated the role of adipocyte PU.1 in the development of the age-associated metabolic syndrome. Methods: We generated mice with adipocyte-specific PU.1 knockout, assessed metabolic changes in young and older adult PU.1fl/fl (control) and AdipoqCre PU.1fl/fl (aPU.1KO) mice, including body weight, body composition, energy expenditure, and glucose homeostasis. We also performed transcriptional analyses using RNA-Sequencing of adipocytes from these mice. Results: aPU.1KO mice have elevated energy expenditure at a young age and decreased adiposity and increased insulin sensitivity in later life. Corroborating these observations, transcriptional network analysis indicated the existence of validated, adipocyte PU.1-modulated regulatory hubs that direct inflammatory and thermogenic gene expression programs. Conclusion: Our data provide evidence for a previously uncharacterized role of PU.1 in the development of age-associated obesity and insulin resistance.
Collapse
Affiliation(s)
- Ke Yun Chen
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX, United States
| | - Alejandra De Angulo
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX, United States
| | - Xin Guo
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX, United States
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Aditya More
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX, United States
| | - Scott A. Ochsner
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Eduardo Lopez
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX, United States
| | - David Saul
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX, United States
| | - Weijun Pang
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX, United States
- Northwestern University of Agriculture and Forestry, Yangling, China
| | - Yuxiang Sun
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX, United States
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| | - Neil J. McKenna
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- *Correspondence: Neil J. McKenna, ; Qiang Tong,
| | - Qiang Tong
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX, United States
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Huffington Center on Aging, Houston, TX, United States
- Department of Medicine, Baylor College of Medicine, Huffington Center on Aging, Houston, TX, United States
- *Correspondence: Neil J. McKenna, ; Qiang Tong,
| |
Collapse
|
34
|
Nuclear Receptors in Energy Metabolism. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1390:61-82. [DOI: 10.1007/978-3-031-11836-4_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
35
|
Adipocyte Biology from the Perspective of In Vivo Research: Review of Key Transcription Factors. Int J Mol Sci 2021; 23:ijms23010322. [PMID: 35008748 PMCID: PMC8745732 DOI: 10.3390/ijms23010322] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 12/14/2022] Open
Abstract
Obesity and type 2 diabetes are both significant contributors to the contemporary pandemic of non-communicable diseases. Both disorders are interconnected and associated with the disruption of normal homeostasis in adipose tissue. Consequently, exploring adipose tissue differentiation and homeostasis is important for the treatment and prevention of metabolic disorders. The aim of this work is to review the consecutive steps in the postnatal development of adipocytes, with a special emphasis on in vivo studies. We gave particular attention to well-known transcription factors that had been thoroughly described in vitro, and showed that the in vivo research of adipogenic differentiation can lead to surprising findings.
Collapse
|
36
|
Cataldi S, Aprile M, Melillo D, Mucel I, Giorgetti-Peraldi S, Cormont M, Italiani P, Blüher M, Tanti JF, Ciccodicola A, Costa V. TNFα Mediates Inflammation-Induced Effects on PPARG Splicing in Adipose Tissue and Mesenchymal Precursor Cells. Cells 2021; 11:cells11010042. [PMID: 35011604 PMCID: PMC8750445 DOI: 10.3390/cells11010042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 12/16/2021] [Accepted: 12/20/2021] [Indexed: 01/18/2023] Open
Abstract
Low-grade chronic inflammation and reduced differentiation capacity are hallmarks of hypertrophic adipose tissue (AT) and key contributors of insulin resistance. We identified PPARGΔ5 as a dominant-negative splicing isoform overexpressed in the AT of obese/diabetic patients able to impair adipocyte differentiation and PPARγ activity in hypertrophic adipocytes. Herein, we investigate the impact of macrophage-secreted pro-inflammatory factors on PPARG splicing, focusing on PPARGΔ5. We report that the epididymal AT of LPS-treated mice displays increased PpargΔ5/cPparg ratio and reduced expression of Pparg-regulated genes. Interestingly, pro-inflammatory factors secreted from murine and human pro-inflammatory macrophages enhance the PPARGΔ5/cPPARG ratio in exposed adipogenic precursors. TNFα is identified herein as factor able to alter PPARG splicing—increasing PPARGΔ5/cPPARG ratio—through PI3K/Akt signaling and SRp40 splicing factor. In line with in vitro data, TNFA expression is higher in the SAT of obese (vs. lean) patients and positively correlates with PPARGΔ5 levels. In conclusion, our results indicate that inflammatory factors secreted by metabolically-activated macrophages are potent stimuli that modulate the expression and splicing of PPARG. The resulting imbalance between canonical and dominant negative isoforms may crucially contribute to impair PPARγ activity in hypertrophic AT, exacerbating the defective adipogenic capacity of precursor cells.
Collapse
Affiliation(s)
- Simona Cataldi
- Institute of Genetics and Biophysics ‘‘Adriano Buzzati-Traverso’’, CNR, Via P. Castellino 111, 80131 Naples, Italy; (S.C.); (M.A.); (A.C.)
| | - Marianna Aprile
- Institute of Genetics and Biophysics ‘‘Adriano Buzzati-Traverso’’, CNR, Via P. Castellino 111, 80131 Naples, Italy; (S.C.); (M.A.); (A.C.)
| | - Daniela Melillo
- Institute of Biochemistry and Cell Biology, CNR, Via P. Castellino 111, 80131 Naples, Italy; (D.M.); (P.I.)
| | - Inès Mucel
- Université Côte d’Azur, Inserm UMR1065, C3M, Team Cellular and Molecular Pathophysiology of Obesity, 06204 Nice, France; (I.M.); (S.G.-P.); (M.C.); (J.-F.T.)
| | - Sophie Giorgetti-Peraldi
- Université Côte d’Azur, Inserm UMR1065, C3M, Team Cellular and Molecular Pathophysiology of Obesity, 06204 Nice, France; (I.M.); (S.G.-P.); (M.C.); (J.-F.T.)
| | - Mireille Cormont
- Université Côte d’Azur, Inserm UMR1065, C3M, Team Cellular and Molecular Pathophysiology of Obesity, 06204 Nice, France; (I.M.); (S.G.-P.); (M.C.); (J.-F.T.)
| | - Paola Italiani
- Institute of Biochemistry and Cell Biology, CNR, Via P. Castellino 111, 80131 Naples, Italy; (D.M.); (P.I.)
| | - Matthias Blüher
- Medical Department III-Endocrinology, Nephrology and Rheumatology, University of Leipzig, 04103 Leipzig, Germany;
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Philipp-Rosenthal-Str. 27, 04103 Leipzig, Germany
| | - Jean-François Tanti
- Université Côte d’Azur, Inserm UMR1065, C3M, Team Cellular and Molecular Pathophysiology of Obesity, 06204 Nice, France; (I.M.); (S.G.-P.); (M.C.); (J.-F.T.)
| | - Alfredo Ciccodicola
- Institute of Genetics and Biophysics ‘‘Adriano Buzzati-Traverso’’, CNR, Via P. Castellino 111, 80131 Naples, Italy; (S.C.); (M.A.); (A.C.)
- Department of Science and Technology, University of Naples ‘‘Parthenope’’, 80143 Naples, Italy
| | - Valerio Costa
- Institute of Genetics and Biophysics ‘‘Adriano Buzzati-Traverso’’, CNR, Via P. Castellino 111, 80131 Naples, Italy; (S.C.); (M.A.); (A.C.)
- Correspondence: ; Tel.: +39-0816132617
| |
Collapse
|
37
|
Casado-Díaz A, Rodríguez-Ramos Á, Torrecillas-Baena B, Dorado G, Quesada-Gómez JM, Gálvez-Moreno MÁ. Flavonoid Phloretin Inhibits Adipogenesis and Increases OPG Expression in Adipocytes Derived from Human Bone-Marrow Mesenchymal Stromal-Cells. Nutrients 2021; 13:4185. [PMID: 34836440 PMCID: PMC8623874 DOI: 10.3390/nu13114185] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/20/2022] Open
Abstract
Phloretin (a flavonoid abundant in apple), has antioxidant, anti-inflammatory, and glucose-transporter inhibitory properties. Thus, it has interesting pharmacological and nutraceutical potential. Bone-marrow mesenchymal stem cells (MSC) have high differentiation capacity, being essential for maintaining homeostasis and regenerative capacity in the organism. Yet, they preferentially differentiate into adipocytes instead of osteoblasts with aging. This has a negative impact on bone turnover, remodeling, and formation. We have evaluated the effects of phloretin on human adipogenesis, analyzing MSC induced to differentiate into adipocytes. Expression of adipogenic genes, as well as genes encoding OPG and RANKL (involved in osteoclastogenesis), protein synthesis, lipid-droplets formation, and apoptosis, were studied. Results showed that 10 and 20 µM phloretin inhibited adipogenesis. This effect was mediated by increasing beta-catenin, as well as increasing apoptosis in adipocytes, at late stages of differentiation. In addition, this chemical increased OPG gene expression and OPG/RANKL ratio in adipocytes. These results suggest that this flavonoid (including phloretin-rich foods) has interesting potential for clinical and regenerative-medicine applications. Thus, such chemicals could be used to counteract obesity and prevent bone-marrow adiposity. That is particularly useful to protect bone mass and treat diseases like osteoporosis, which is an epidemic worldwide.
Collapse
Affiliation(s)
- Antonio Casado-Díaz
- Unidad de Gestión Clínica de Endocrinología y Nutrición—GC17, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, CIBERFES, 14004 Córdoba, Spain; (Á.R.-R.); (B.T.-B.); (J.M.Q.-G.); (M.Á.G.-M.)
| | - Ángel Rodríguez-Ramos
- Unidad de Gestión Clínica de Endocrinología y Nutrición—GC17, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, CIBERFES, 14004 Córdoba, Spain; (Á.R.-R.); (B.T.-B.); (J.M.Q.-G.); (M.Á.G.-M.)
| | - Bárbara Torrecillas-Baena
- Unidad de Gestión Clínica de Endocrinología y Nutrición—GC17, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, CIBERFES, 14004 Córdoba, Spain; (Á.R.-R.); (B.T.-B.); (J.M.Q.-G.); (M.Á.G.-M.)
| | - Gabriel Dorado
- Dep. Bioquímica y Biología Molecular, Campus Rabanales C6-1-E17, Campus de Excelencia Internacional Agroalimentario (ceiA3), Universidad de Córdoba, CIBERFES, 14071 Córdoba, Spain;
| | - José Manuel Quesada-Gómez
- Unidad de Gestión Clínica de Endocrinología y Nutrición—GC17, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, CIBERFES, 14004 Córdoba, Spain; (Á.R.-R.); (B.T.-B.); (J.M.Q.-G.); (M.Á.G.-M.)
| | - María Ángeles Gálvez-Moreno
- Unidad de Gestión Clínica de Endocrinología y Nutrición—GC17, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, CIBERFES, 14004 Córdoba, Spain; (Á.R.-R.); (B.T.-B.); (J.M.Q.-G.); (M.Á.G.-M.)
| |
Collapse
|
38
|
Chen H, Gao H, Xie HT, Liu ST, Huang YK, Zhang MC. Hyperkeratinization and Proinflammatory Cytokine Expression in Meibomian Glands Induced by Staphylococcus aureus. Invest Ophthalmol Vis Sci 2021; 62:11. [PMID: 34643663 PMCID: PMC8525831 DOI: 10.1167/iovs.62.13.11] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 09/15/2021] [Indexed: 02/06/2023] Open
Abstract
Purpose This exploratory study aimed to investigate the morphological and pathological alterations of the meibomian gland (MG) with the Staphylococcus aureus crude extracts (SACEs) treatment. Methods Mouse MG explants were cultured and differentiated with or without SACEs for 48 hours. Explant's viability and cell death were determined by thiazolyl blue tetrazolium bromide (MTT) assay and TUNEL assay. MG morphology was observed by Hematoxylin and Eosin staining. Lipid droplet production was detected by Nile Red staining and LipidTox immunostaining. The pro-inflammatory cytokines were detected by ELISA. The relative gene and protein expression in MG explants was determined via quantitative RT-PCR, immunostaining, and immunoblotting. The components of the SACEs were analyzed by immunoblotting and silver staining. Results Our findings demonstrated that the SACEs treatment induced overexpression of keratin 1 (Krt1) in the ducts and acini of MG explants, accompanied by a decrease in viability and an increase in cell death in explants. Furthermore, the SACEs treatment dose-dependently increased the levels of TNF-α, IL-1β, and IL-6 in MG explants. The SACEs treatment induced activation of the nuclear factor kappa B (NF-κB) and AIM2 (absent in melanoma 2)/ASC (apoptosis-associated speck-like protein containing a caspase recruitment domain) inflammasome signaling pathway in explants. Further investigation showed expression of the key adipogenesis-related molecule peroxisome proliferator-activated receptor γ was decreased after SACEs treatment. However, no change was found in the lipid synthesis of MG explants after treatment with the SACEs. Staphylococcal enterotoxins B (SEB) was detected in the SACEs. SEB induced the overexpression of Krt1 and IL-1β in ducts and acini of MG explants. Conclusions Our findings confirm that Staphylococcus aureus induced hyperkeratinization and pro-inflammatory cytokines expression in MG explants ducts and acini. These effects might be mediated by SEB. Activation of the NF-κB and AIM2/ASC signaling pathway is involved in this process.
Collapse
Affiliation(s)
- Hua Chen
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Gao
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hua-Tao Xie
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shu-Ting Liu
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Kan Huang
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming-Chang Zhang
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
39
|
Hikage F, Ichioka H, Watanabe M, Umetsu A, Ohguro H, Ida Y. Addition of ROCK inhibitors to prostaglandin derivative (PG) synergistically affects adipogenesis of the 3D spheroids of human orbital fibroblasts (HOFs). Hum Cell 2021; 35:125-132. [PMID: 34591280 DOI: 10.1007/s13577-021-00623-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 09/27/2021] [Indexed: 11/30/2022]
Abstract
To study the additive effects of Rho-associated coiled-coil containing protein kinase inhibitors, ripasudil (Rip) to bimatoprost acid (BIM-A) on orbital adipose tissue, three-dimensional (3D) cultures of human orbital fibroblasts (HOFs) were prepared and the physical properties including the 3D spheroid size and stiffness, lipid staining by BODIPY and the mRNA expression of adipogenesis-related genes, PPARγ and AP2, and extracellular matrix (ECM) including collagen (COL)1, 4 and 6, and fibronectin (FN) were analyzed. Adipogenesis (DIF+) induced (1) enlargement and increasing stiffness of the 3D HOFs spheroid, (2) increased lipid staining, the expression of adipogenesis-related gene expressions, and (3) the down-regulation of COL1 and FN and up-regulation of COL4 and COL6. In the presence of BIM-A, (1) such DIF+-induced changes in 3D spheroid size and stiffness were significantly inhibited or enhanced, respectively, (2) the lipid staining and its related gene expressions were significantly down-regulated, and (3) the expression of COL1 and COL6 were up-regulated. By the addition of Rip to BIM-A, the above BIM-A-induced effects were all inhibited, except for the up-regulation of COL6 and FN expression, that is, enlarging and decreasing stiffness, enhancement of lipid staining and its related gene expression, and down-regulation of COL1 expression. Our present study indicates that Rip significantly suppressed BIM-A-induced effects toward 3D HOFs spheroids.
Collapse
Affiliation(s)
- Fumihito Hikage
- Departments of Ophthalmology, Sapporo Medical University School of Medicine, Sapporo, Hokkaido, Japan
| | - Hanae Ichioka
- Departments of Ophthalmology, Sapporo Medical University School of Medicine, Sapporo, Hokkaido, Japan
| | - Megumi Watanabe
- Departments of Ophthalmology, Sapporo Medical University School of Medicine, Sapporo, Hokkaido, Japan
| | - Araya Umetsu
- Departments of Ophthalmology, Sapporo Medical University School of Medicine, Sapporo, Hokkaido, Japan
| | - Hiroshi Ohguro
- Departments of Ophthalmology, Sapporo Medical University School of Medicine, Sapporo, Hokkaido, Japan
| | - Yosuke Ida
- Departments of Ophthalmology, Sapporo Medical University School of Medicine, Sapporo, Hokkaido, Japan.
| |
Collapse
|
40
|
PARPs in lipid metabolism and related diseases. Prog Lipid Res 2021; 84:101117. [PMID: 34450194 DOI: 10.1016/j.plipres.2021.101117] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/10/2021] [Accepted: 08/18/2021] [Indexed: 12/28/2022]
Abstract
PARPs and tankyrases (TNKS) represent a family of 17 proteins. PARPs and tankyrases were originally identified as DNA repair factors, nevertheless, recent advances have shed light on their role in lipid metabolism. To date, PARP1, PARP2, PARP3, tankyrases, PARP9, PARP10, PARP14 were reported to have multi-pronged connections to lipid metabolism. The activity of PARP enzymes is fine-tuned by a set of cholesterol-based compounds as oxidized cholesterol derivatives, steroid hormones or bile acids. In turn, PARPs modulate several key processes of lipid homeostasis (lipotoxicity, fatty acid and steroid biosynthesis, lipoprotein homeostasis, fatty acid oxidation, etc.). PARPs are also cofactors of lipid-responsive nuclear receptors and transcription factors through which PARPs regulate lipid metabolism and lipid homeostasis. PARP activation often represents a disruptive signal to (lipid) metabolism, and PARP-dependent changes to lipid metabolism have pathophysiological role in the development of hyperlipidemia, obesity, alcoholic and non-alcoholic fatty liver disease, type II diabetes and its complications, atherosclerosis, cardiovascular aging and skin pathologies, just to name a few. In this synopsis we will review the evidence supporting the beneficial effects of pharmacological PARP inhibitors in these diseases/pathologies and propose repurposing PARP inhibitors already available for the treatment of various malignancies.
Collapse
|
41
|
PPARs-Orchestrated Metabolic Homeostasis in the Adipose Tissue. Int J Mol Sci 2021; 22:ijms22168974. [PMID: 34445679 PMCID: PMC8396609 DOI: 10.3390/ijms22168974] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/17/2021] [Accepted: 08/17/2021] [Indexed: 01/12/2023] Open
Abstract
It has been more than three decades since peroxisome proliferator-activated receptors (PPARs) were first discovered. Many investigations have revealed the central regulators of PPARs in lipid and glucose homeostasis in response to different nutrient conditions. PPARs have attracted much attention due to their ability to improve metabolic syndromes, and they have also been proposed as classical drug targets for the treatment of hyperlipidemia and type 2 diabetes (T2D) mellitus. In parallel, adipose tissue is known to play a unique role in the pathogenesis of insulin resistance and metabolic syndromes due to its ability to “safely” store lipids and secrete cytokines that regulate whole-body metabolism. Adipose tissue relies on a complex and subtle network of transcription factors to maintain its normal physiological function, by coordinating various molecular events, among which PPARs play distinctive and indispensable roles in adipocyte differentiation, lipid metabolism, adipokine secretion, and insulin sensitivity. In this review, we discuss the characteristics of PPARs with special emphasis on the roles of the different isotypes in adipocyte biology.
Collapse
|
42
|
Rapuano R, Ziccardi P, Cioffi V, Dallavalle S, Moricca S, Lupo A. Cladosporols A and B, two natural peroxisome proliferator-activated receptor gamma (PPARγ) agonists, inhibit adipogenesis in 3T3-L1 preadipocytes and cause a conditioned-culture-medium-dependent arrest of HT-29 cell proliferation. Biochim Biophys Acta Gen Subj 2021; 1865:129973. [PMID: 34352342 DOI: 10.1016/j.bbagen.2021.129973] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/12/2021] [Accepted: 07/26/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Obesity and type 2 diabetes mellitus, which are widespread throughout the world, require therapeutic interventions targeted to solve clinical problems (insulin resistance, hyperglycaemia, dyslipidaemia and steatosis). Several natural compounds are now part of the therapeutic repertoire developed to better manage these pathological conditions. Cladosporols, secondary metabolites from the fungus Cladosporium tenuissimum, have been characterised for their ability to control cell proliferation in human colon cancer cell lines through peroxisome proliferator-activated receptor gamma (PPARγ)-mediated modulation of gene expression. Here, we report data concerning the ability of cladosporols to regulate the differentiation of murine 3T3-L1 preadipocytes. METHODS Cell counting and MTT assay were used for analysing cell proliferation. RT-PCR and Western blotting assays were performed to evaluate differentiation marker expression. Cell migration was analysed by wound-healing assay. RESULTS We showed that cladosporol A and B inhibited the storage of lipids in 3T3-L1 mature adipocytes, while their administration did not affect the proliferative ability of preadipocytes. Moreover, both cladosporols downregulated mRNA and protein levels of early (C/EBPα and PPARγ) and late (aP2, LPL, FASN, GLUT-4, adiponectin and leptin) differentiation markers of adipogenesis. Finally, we found that proliferation and migration of HT-29 colorectal cancer cells were inhibited by conditioned medium from cladosporol-treated 3T3-L1 cells compared with the preadipocyte conditioned medium. CONCLUSIONS To our knowledge, this is the first report describing that cladosporols inhibit in vitro adipogenesis and through this inhibition may interfere with HT-29 cancer cell growth and migration. GENERAL SIGNIFICANCE Cladosporols are promising tools to inhibit concomitantly adipogenesis and control colon cancer initiation and progression.
Collapse
Affiliation(s)
- Roberta Rapuano
- Dipartimento di Scienze e Tecnologie, Università del Sannio, via Port'Arsa 11, 82100 Benevento, Italy
| | - Pamela Ziccardi
- Dipartimento di Scienze e Tecnologie, Università del Sannio, via Port'Arsa 11, 82100 Benevento, Italy
| | - Valentina Cioffi
- Dipartimento di Scienze e Tecnologie, Università del Sannio, via Port'Arsa 11, 82100 Benevento, Italy
| | - Sabrina Dallavalle
- Dipartimento di Scienze per gli Alimenti, la Nutrizione e l'Ambiente, Università degli Studi di Milano, via Celoria 2, 20133 Milano, Italy
| | - Salvatore Moricca
- Dipartimento di Scienze delle Produzioni Agroalimentari e dell'Ambiente (DiSPAA), Università degli Studi di Firenze, Piazzale delle Cascine 18, 50144 Firenze, Italy
| | - Angelo Lupo
- Dipartimento di Scienze e Tecnologie, Università del Sannio, via Port'Arsa 11, 82100 Benevento, Italy.
| |
Collapse
|
43
|
Kubota Y, Nagano H, Kosaka K, Ogata H, Nakayama A, Yokoyama M, Murata K, Akita S, Kuriyama M, Furuyama N, Kuroda M, Tanaka T, Mitsukawa N. Epigenetic modifications underlie the differential adipogenic potential of preadipocytes derived from human subcutaneous fat tissue. Am J Physiol Cell Physiol 2021; 321:C596-C606. [PMID: 34319829 DOI: 10.1152/ajpcell.00387.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
AIM Ceiling culture-derived preadipocytes (ccdPAs) and adipose-derived stem cells (ASCs) can be harvested from human subcutaneous fat tissue using the specific gravity method. Both cell types possess a similar spindle shape without lipid droplets. We previously reported that ccdPAs have a higher adipogenic potential than ASCs, even after a 7-week culture. We performed a genome-wide epigenetic analysis to examine the mechanisms contributing to the adipogenic potential differences between ccdPAs and ASCs. MATERIALS AND METHODS Methylation analysis of cytosines followed by guanine (CpG) using a 450K BeadChip was performed on human ccdPAs and ASCs isolated from three metabolically healthy females. Chromatin immunoprecipitation sequencing (ChIP-seq) was performed to evaluate trimethylation at lysine 4 of histone 3 (H3K4me3). RESULTS Unsupervised machine learning using t-distributed stochastic neighbor embedding (tSNE) to interpret 450,000-dimensional methylation assay data showed that the cells were divided into ASC and ccdPA groups. In KEGG pathway analysis of 1,543 genes with differential promoter CpG methylation, the peroxisome proliferator-activated receptor (PPAR) and adipocytokine signaling pathways ranked in the top 10 pathways. In the PPAR gamma gene, H3K4me3 peak levels were higher in ccdPAs than in ASCs, whereas promoter CpG methylation levels were significantly lower in ccdPAs than in ASCs. Similar differences in promoter CpG methylation were also seen in the fatty acid-binding protein 4 (FABP4) and leptin genes. CONCLUSION We analyzed the epigenetic status of adipogenesis-related genes as a potential mechanism underlying the differences in adipogenic differentiation capability between ASCs and ccdPAs.
Collapse
Affiliation(s)
- Yoshitaka Kubota
- Department of Plastic Surgery, Chiba University, Chiba-city, Chiba, Japan
| | - Hidekazu Nagano
- Department of Molecular Diagnosis, Chiba University, Chiba-city, Chiba, Japan
| | - Kentaro Kosaka
- Department of Plastic Surgery, Chiba University, Chiba-city, Chiba, Japan
| | - Hideyuki Ogata
- Department of Plastic Surgery, Chiba University, Chiba-city, Chiba, Japan
| | - Akitoshi Nakayama
- Department of Molecular Diagnosis, Chiba University, Chiba-city, Chiba, Japan
| | - Masataka Yokoyama
- Department of Molecular Diagnosis, Chiba University, Chiba-city, Chiba, Japan
| | - Kazutaka Murata
- Department of Molecular Diagnosis, Chiba University, Chiba-city, Chiba, Japan
| | - Shinsuke Akita
- Department of Plastic Surgery, Chiba University, Chiba-city, Chiba, Japan
| | - Motone Kuriyama
- Department of Plastic Surgery, Chiba University, Chiba-city, Chiba, Japan
| | | | - Masayuki Kuroda
- Center for Advanced Medicine, Chiba University, Chiba-city, Chiba, Japan
| | - Tomoaki Tanaka
- Department of Molecular Diagnosis, Chiba University, Chiba-city, Chiba, Japan
| | - Nobuyuki Mitsukawa
- Department of Plastic Surgery, Chiba University, Chiba-city, Chiba, Japan
| |
Collapse
|
44
|
Lv X, Qiu J, Hao T, Zhang H, Jiang H, Tan Y. HDAC inhibitor Trichostatin A suppresses adipogenesis in 3T3-L1 preadipocytes. Aging (Albany NY) 2021; 13:17489-17498. [PMID: 34232916 PMCID: PMC8312440 DOI: 10.18632/aging.203238] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/04/2021] [Indexed: 12/30/2022]
Abstract
Background and purpose: Obesity is becoming a major global health issue and is mainly induced by the accumulation of adipose tissues mediated by adipogenesis, which is reported to be regulated by peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT enhancer-binding protein α (C/EBPα). Trichostatin A (TSA) is a novel histone deacetylase inhibitor (HDACI) that was recently reported to exert multiple pharmacological functions. The present study will investigate the inhibitory effect of TSA on adipogenesis, as well as the underlying mechanism. Methods: The adipogenesis of 3T3-L1 cells was induced by stimulation with a differentiation cocktail (DMI) medium for 8 days. MTT assay was used to measure the cell viability and Oil Red O staining was used to evaluate the adipogenesis of 3T3-L1 cells. The total level of triglyceride and released glycerol were detected to evaluate the lipolysis during 3T3-L1 adipogenesis. The expression levels of Leptin, fatty acid-binding protein 4 (FABP4), and sterol regulatory element-binding protein (SREBP1C) were determined by qRT-PCR. qRT-PCR assay was utilized to detect the expression levels of PPARγ and C/EBPα in 3T3-L1 cells. A high-fat diet (HFD) was used to construct an obese mice model, followed by the treatment with TSA. HE staining was conducted to evaluate the pathological state of adipose tissues. Body weights and the weights of adipose tissues were recorded to evaluate the anti-obesity property of TSA. Results: Firstly, the promoted lipid accumulation induced by DMI incubation was significantly reversed by the treatment with TSA in a dose-dependent manner. The elevated expression levels of Leptin, FABP4, SREBP1C, PPARγ, and C/EBPα induced by the stimulation with DMI incubation were dramatically inhibited by the introduction of TSA, accompanied by the upregulation of phosphorylated AMP-activated protein kinase (p-AMPK). Secondly, the inhibitory effect of TSA against the expression level of PPARγ and lipid accumulation was greatly abolished by an AMPK inhibitor. Lastly, the increased body weights and visceral adipocyte tissue weight, as well as the enlarged size of adipocytes induced by HFD were pronouncedly reversed by the administration of TSA. Conclusion: TSA inhibited adipogenesis in 3T3-L1 preadipocytes by activating the AMPK pathway.
Collapse
Affiliation(s)
- Xin Lv
- Department of Clinical Nutrition, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong, China.,Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong, China
| | - Jun Qiu
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong, China
| | - Tao Hao
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong, China
| | - Haoran Zhang
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong, China
| | - Haiping Jiang
- Department of Clinical Nutrition, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong, China.,Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong, China
| | - Yang Tan
- Department of Pathology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518000, Guangdong, China
| |
Collapse
|
45
|
Rauch A, Mandrup S. Transcriptional networks controlling stromal cell differentiation. Nat Rev Mol Cell Biol 2021; 22:465-482. [PMID: 33837369 DOI: 10.1038/s41580-021-00357-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2021] [Indexed: 02/02/2023]
Abstract
Stromal progenitors are found in many different tissues, where they play an important role in the maintenance of tissue homeostasis owing to their ability to differentiate into parenchymal cells. These progenitor cells are differentially pre-programmed by their tissue microenvironment but, when cultured and stimulated in vitro, these cells - commonly referred to as mesenchymal stromal cells (MSCs) - exhibit a marked plasticity to differentiate into many different cell lineages. Loss-of-function studies in vitro and in vivo have uncovered the involvement of specific signalling pathways and key transcriptional regulators that work in a sequential and coordinated fashion to activate lineage-selective gene programmes. Recent advances in omics and single-cell technologies have made it possible to obtain system-wide insights into the gene regulatory networks that drive lineage determination and cell differentiation. These insights have important implications for the understanding of cell differentiation, the contribution of stromal cells to human disease and for the development of cell-based therapeutic applications.
Collapse
Affiliation(s)
- Alexander Rauch
- Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital and Department of Clinical Research, University of Southern Denmark, Odense, Denmark. .,Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark.
| | - Susanne Mandrup
- Center for Functional Genomics and Tissue Plasticity, Functional Genomics & Metabolism Research Unit, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
46
|
Wang Y, Li X, Cao Y, Xiao C, Liu Y, Jin H, Cao Y. Effect of the ACAA1 Gene on Preadipocyte Differentiation in Sheep. Front Genet 2021; 12:649140. [PMID: 34234807 PMCID: PMC8255805 DOI: 10.3389/fgene.2021.649140] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 05/18/2021] [Indexed: 01/01/2023] Open
Abstract
Acetyl-CoA acyltransferase 1 (ACAA1) functions as a key regulator of fatty acid β-oxidation in peroxisomes by catalyzing the cleavage of 3-ketoacyl-CoA to acetyl-CoA and acyl-CoA, which participate in the extension and degradation of fatty acids. Thus, ACAA1 is an important regulator of lipid metabolism and plays an essential role in fatty acid oxidation and lipid metabolism. Our previous study findings revealed that ACAA1 is closely associated with the peroxisome proliferator-activated receptor (PPAR) signaling and fatty acid metabolism pathways, which are involved in fat deposition in sheep, leading to our hypothesis that ACAA1 may be involved in fat deposition by regulating lipid metabolism. However, the associated molecular mechanism remains unclear. In the present study, to assess the potential function of ACAA1 in sheep preadipocyte differentiation, we knocked down and overexpressed ACAA1 in sheep preadipocytes and evaluated the pattern of ACAA1 gene expression during preadipocyte differentiation by qRT-PCR. ACAA1 was significantly expressed in the early stage of adipocyte differentiation, and then its expression decreased. ACAA1 deficiency increased lipid accumulation and the triglyceride content and promoted sheep preadipocyte differentiation, whereas ACAA1 overexpression inhibited adipogenesis and decreased lipid accumulation and the triglyceride content. Simultaneously, we demonstrated that ACAA1 deficiency upregulated the expressions of the adipogenic marker genes PPARγ and C/EBPα in sheep preadipocytes, but ACAA1 overexpression inhibited the expressions of these markers, indicating that ACAA1 affects lipid metabolism by regulating adipogenic marker genes. Our results may promote a better understanding of the regulation of adipogenesis by ACAA1.
Collapse
Affiliation(s)
- Yanli Wang
- Institute of Animal Biotechnology, Jilin Academy of Agricultural Science, Changchun, China
| | - Xin Li
- Institute of Animal Biotechnology, Jilin Academy of Agricultural Science, Changchun, China
| | - Yang Cao
- Institute of Animal Husbandry and Veterinary, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Cheng Xiao
- Institute of Animal Biotechnology, Jilin Academy of Agricultural Science, Changchun, China
| | - Yu Liu
- Institute of Animal Biotechnology, Jilin Academy of Agricultural Science, Changchun, China
| | - Haiguo Jin
- Institute of Animal Biotechnology, Jilin Academy of Agricultural Science, Changchun, China
| | - Yang Cao
- Institute of Animal Biotechnology, Jilin Academy of Agricultural Science, Changchun, China
| |
Collapse
|
47
|
Wagner G, Fenzl A, Lindroos-Christensen J, Einwallner E, Husa J, Witzeneder N, Rauscher S, Gröger M, Derdak S, Mohr T, Sutterlüty H, Klinglmüller F, Wolkerstorfer S, Fondi M, Hoermann G, Cao L, Wagner O, Kiefer FW, Esterbauer H, Bilban M. LMO3 reprograms visceral adipocyte metabolism during obesity. J Mol Med (Berl) 2021; 99:1151-1171. [PMID: 34018016 PMCID: PMC8313462 DOI: 10.1007/s00109-021-02089-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 05/05/2021] [Accepted: 05/10/2021] [Indexed: 01/02/2023]
Abstract
Abstract Obesity and body fat distribution are important risk factors for the development of type 2 diabetes and metabolic syndrome. Evidence has accumulated that this risk is related to intrinsic differences in behavior of adipocytes in different fat depots. We recently identified LIM domain only 3 (LMO3) in human mature visceral adipocytes; however, its function in these cells is currently unknown. The aim of this study was to determine the potential involvement of LMO3-dependent pathways in the modulation of key functions of mature adipocytes during obesity. Based on a recently engineered hybrid rAAV serotype Rec2 shown to efficiently transduce both brown adipose tissue (BAT) and white adipose tissue (WAT), we delivered YFP or Lmo3 to epididymal WAT (eWAT) of C57Bl6/J mice on a high-fat diet (HFD). The effects of eWAT transduction on metabolic parameters were evaluated 10 weeks later. To further define the role of LMO3 in insulin-stimulated glucose uptake, insulin signaling, adipocyte bioenergetics, as well as endocrine function, experiments were conducted in 3T3-L1 adipocytes and newly differentiated human primary mature adipocytes, engineered for transient gain or loss of LMO3 expression, respectively. AAV transduction of eWAT results in strong and stable Lmo3 expression specifically in the adipocyte fraction over a course of 10 weeks with HFD feeding. LMO3 expression in eWAT significantly improved insulin sensitivity and healthy visceral adipose tissue expansion in diet-induced obesity, paralleled by increased serum adiponectin. In vitro, LMO3 expression in 3T3-L1 adipocytes increased PPARγ transcriptional activity, insulin-stimulated GLUT4 translocation and glucose uptake, as well as mitochondrial oxidative capacity in addition to fatty acid oxidation. Mechanistically, LMO3 induced the PPARγ coregulator Ncoa1, which was required for LMO3 to enhance glucose uptake and mitochondrial oxidative gene expression. In human mature adipocytes, LMO3 overexpression promoted, while silencing of LMO3 suppressed mitochondrial oxidative capacity. LMO3 expression in visceral adipose tissue regulates multiple genes that preserve adipose tissue functionality during obesity, such as glucose metabolism, insulin sensitivity, mitochondrial function, and adiponectin secretion. Together with increased PPARγ activity and Ncoa1 expression, these gene expression changes promote insulin-induced GLUT4 translocation, glucose uptake in addition to increased mitochondrial oxidative capacity, limiting HFD-induced adipose dysfunction. These data add LMO3 as a novel regulator improving visceral adipose tissue function during obesity. Key messages
LMO3 increases beneficial visceral adipose tissue expansion and insulin sensitivity in vivo. LMO3 increases glucose uptake and oxidative mitochondrial activity in adipocytes. LMO3 increases nuclear coactivator 1 (Ncoa1). LMO3-enhanced glucose uptake and mitochondrial gene expression requires Ncoa1. Supplementary Information The online version contains supplementary material available at 10.1007/s00109-021-02089-9.
Collapse
Affiliation(s)
- Gabriel Wagner
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Anna Fenzl
- Clinical Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, 1090, Vienna, Austria
| | - Josefine Lindroos-Christensen
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria.,Novo Nordisk, Maaloev, Denmark
| | - Elisa Einwallner
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Julia Husa
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Nadine Witzeneder
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Sabine Rauscher
- Core Facilities, Medical University of Vienna, 1090, Vienna, Austria
| | - Marion Gröger
- Core Facilities, Medical University of Vienna, 1090, Vienna, Austria
| | - Sophia Derdak
- Core Facilities, Medical University of Vienna, 1090, Vienna, Austria
| | - Thomas Mohr
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Hedwig Sutterlüty
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Florian Klinglmüller
- Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, 1090, Vienna, Austria.,Austrian Medicines & Medical Devices Agency, 1200, Vienna, Austria
| | - Silviya Wolkerstorfer
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria.,University of Applied Sciences, FH Campus Wien, 1100, Vienna, Austria.,Institute of Cardiovascular Prevention, Ludwig-Maximilians-University, 80336, Munich, Germany
| | - Martina Fondi
- University of Applied Sciences, FH Campus Wien, 1100, Vienna, Austria
| | - Gregor Hoermann
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria.,Central Institute of Medical and Chemical Laboratory Diagnostics, University Hospital Innsbruck, 6020, Innsbruck, Austria
| | - Lei Cao
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, 43210, USA
| | - Oswald Wagner
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Florian W Kiefer
- Clinical Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, 1090, Vienna, Austria
| | - Harald Esterbauer
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Martin Bilban
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria. .,Core Facilities, Medical University of Vienna, 1090, Vienna, Austria.
| |
Collapse
|
48
|
Ercolano G, Gomez-Cadena A, Dumauthioz N, Vanoni G, Kreutzfeldt M, Wyss T, Michalik L, Loyon R, Ianaro A, Ho PC, Borg C, Kopf M, Merkler D, Krebs P, Romero P, Trabanelli S, Jandus C. PPARɣ drives IL-33-dependent ILC2 pro-tumoral functions. Nat Commun 2021; 12:2538. [PMID: 33953160 PMCID: PMC8100153 DOI: 10.1038/s41467-021-22764-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 03/25/2021] [Indexed: 01/27/2023] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) play a critical role in protection against helminths and in diverse inflammatory diseases by responding to soluble factors such as the alarmin IL-33, that is often overexpressed in cancer. Nonetheless, regulatory factors that dictate ILC2 functions remain poorly studied. Here, we show that peroxisome proliferator-activated receptor gamma (PPARγ) is selectively expressed in ILC2s in humans and in mice, acting as a central functional regulator. Pharmacologic inhibition or genetic deletion of PPARγ in ILC2s significantly impair IL-33-induced Type-2 cytokine production and mitochondrial fitness. Further, PPARγ blockade in ILC2s disrupts their pro-tumoral effect induced by IL-33-secreting cancer cells. Lastly, genetic ablation of PPARγ in ILC2s significantly suppresses tumor growth in vivo. Our findings highlight a crucial role for PPARγ in supporting the IL-33 dependent pro-tumorigenic role of ILC2s and suggest that PPARγ can be considered as a druggable pathway in ILC2s to inhibit their effector functions. Hence, PPARγ targeting might be exploited in cancer immunotherapy and in other ILC2-driven mediated disorders, such as asthma and allergy.
Collapse
Affiliation(s)
- Giuseppe Ercolano
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland.,Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
| | - Alejandra Gomez-Cadena
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland.,Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
| | - Nina Dumauthioz
- Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland
| | - Giulia Vanoni
- Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland
| | - Mario Kreutzfeldt
- Department of Pathology and Immunology, Division of Clinical Pathology, University and University Hospitals of Geneva, Geneva, Switzerland
| | - Tania Wyss
- Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland
| | - Liliane Michalik
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Romain Loyon
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France.,University Hospital of Besançon, Department of Medical Oncology, Besançon, France
| | - Angela Ianaro
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Ping-Chih Ho
- Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland
| | - Christophe Borg
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France.,University Hospital of Besançon, Department of Medical Oncology, Besançon, France
| | - Manfred Kopf
- Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland
| | - Doron Merkler
- Department of Pathology and Immunology, Division of Clinical Pathology, University and University Hospitals of Geneva, Geneva, Switzerland
| | - Philippe Krebs
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Pedro Romero
- Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland
| | - Sara Trabanelli
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland.,Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
| | - Camilla Jandus
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland. .,Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland.
| |
Collapse
|
49
|
Okreglicka K, Iten I, Pohlmeier L, Onder L, Feng Q, Kurrer M, Ludewig B, Nielsen P, Schneider C, Kopf M. PPARγ is essential for the development of bone marrow erythroblastic island macrophages and splenic red pulp macrophages. J Exp Med 2021; 218:e20191314. [PMID: 33765133 PMCID: PMC8006858 DOI: 10.1084/jem.20191314] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 12/09/2020] [Accepted: 02/22/2021] [Indexed: 12/13/2022] Open
Abstract
Tissue-resident macrophages play a crucial role in maintaining homeostasis. Macrophage progenitors migrate to tissues perinatally, where environmental cues shape their identity and unique functions. Here, we show that the absence of PPARγ affects neonatal development and VCAM-1 expression of splenic iron-recycling red pulp macrophages (RPMs) and bone marrow erythroblastic island macrophages (EIMs). Transcriptome analysis of the few remaining Pparg-deficient RPM-like and EIM-like cells suggests that PPARγ is required for RPM and EIM identity, cell cycling, migration, and localization, but not function in mature RPMs. Notably, Spi-C, another transcription factor implicated in RPM development, was not essential for neonatal expansion of RPMs, even though the transcriptome of Spic-deficient RPMs was strongly affected and indicated a loss of identity. Similarities shared by Pparg- and Spic-deficient RPM-like cells allowed us to identify pathways that rely on both factors. PPARγ and Spi-C collaborate in inducing transcriptional changes, including VCAM-1 and integrin αD expression, which could be required for progenitor retention in the tissue, allowing access to niche-related signals that finalize differentiation.
Collapse
Affiliation(s)
- Katarzyna Okreglicka
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Irina Iten
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Lea Pohlmeier
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Lucas Onder
- Institute of Immunobiology, Cantonal Hospital, St. Gallen, Switzerland
| | - Qian Feng
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology, Zurich, Switzerland
| | | | - Burkhard Ludewig
- Institute of Immunobiology, Cantonal Hospital, St. Gallen, Switzerland
| | - Peter Nielsen
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Christoph Schneider
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology, Zurich, Switzerland
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Manfred Kopf
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology, Zurich, Switzerland
| |
Collapse
|
50
|
Funk MI, Conde MA, Piwien-Pilipuk G, Uranga RM. Novel antiadipogenic effect of menadione in 3T3-L1 cells. Chem Biol Interact 2021; 343:109491. [PMID: 33945810 DOI: 10.1016/j.cbi.2021.109491] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/31/2021] [Accepted: 04/19/2021] [Indexed: 12/30/2022]
Abstract
Inhibition of adipocyte differentiation can be used as a strategy for preventing adipose tissue expansion and, consequently, for obesity management. Since reactive oxygen species (ROS) have emerged as key modulators of adipogenesis, the effect of menadione (a synthetic form of vitamin K known to induce the increase of intracellular ROS) on 3T3-L1 preadipocyte differentiation was studied. Menadione (15 μM) increased ROS and lipid peroxidation, generating mild oxidative stress without affecting cell viability. Menadione drastically inhibited adipogenesis, accompanied by decreased intracellular lipid accumulation and diminished expression of the lipo/adipogenic markers peroxisome proliferator-activated receptor (PPAR)γ, fatty acid synthase (FAS), CCAAT/enhancer-binding protein (C/EBP) α, fatty acid binding protein (FABP) 4, and perilipin. Menadione treatment also increased lipolysis, as indicated by augmented glycerol release and reinforced by the increased expression of hormone-sensitive lipase (HSL). Additionally, menadione increased the inhibitory phosphorylation of acetyl-CoA-carboxylase (ACC), which results in the inhibition of fatty acid synthesis. As a consequence, triglyceride content was decreased. Menadione also inhibited the phosphatidylinositol 3-kinase (PI3K)/Akt pathway. Further, treatment with increased concentration of insulin, a potent physiological activator of the PI3K/Akt pathway, rescued the normal level of expression of PPARγ, the master regulator of adipogenesis, and overcame the restraining effect of menadione on the differentiation capacity of 3T3-L1 preadipocytes. Our study reveals novel antiadipogenic action for menadione, which is, at least in part, mediated by the PI3K/Akt pathway signaling and raises its potential as a therapeutic agent in the treatment or prevention of adiposity.
Collapse
Affiliation(s)
- Melania Iara Funk
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Universidad Nacional Del Sur (UNS)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina. Departamento de Biología, Bioquímica y Farmacia, UNS, Bahía Blanca, Argentina
| | - Melisa Ailén Conde
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Universidad Nacional Del Sur (UNS)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina. Departamento de Biología, Bioquímica y Farmacia, UNS, Bahía Blanca, Argentina
| | | | - Romina María Uranga
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Universidad Nacional Del Sur (UNS)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina. Departamento de Biología, Bioquímica y Farmacia, UNS, Bahía Blanca, Argentina.
| |
Collapse
|