1
|
Çil EN, Soysal Y. Anti-Obesity Effects of Calcium Fructoborate by Inhibiting Adipogenesis and Increasing SIRT's Expression in 3T3-L1 Cells. Biol Trace Elem Res 2025; 203:3833-3844. [PMID: 39531139 DOI: 10.1007/s12011-024-04444-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Obesity is a global public health problem that can lead to mortality and morbidity. Studies on the pathophysiology of obesity for effective and safe treatments are focused on the mechanisms of adipogenesis. The association between boron treatment and weight loss has been reported, but its anti-adipogenic mechanisms and effects on preadipocytes remain unclear. This study aims to investigate the effects of boron compounds boric acid (BA) and calcium fructoborate (CaFB) on adipogenesis using the most widely used in vitro 3T3-L1 cellular model. In our study, cytotoxicity, Oil Red O (ORO), gene and protein expression analyses and cellular NAD measurements of boron compounds were performed. Peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT/enhancer binding protein α (C/EBPα) transcription factors are the main regulators of adipogenesis, and boron compounds affect them at gene and protein levels by showing anti-obesity effects. This is the first study to show that CaFB has anti-obesity properties in mouse adipocytes. Sirtuins, known as the longevity genes, were also activated from boron treatment. Results of this research provide new basic knowledge and insights into the effect of boron-based compounds on obesity. It also offers potential prospects for the development of effective treatment and/or supportive treatment methods.
Collapse
Affiliation(s)
- Ezgi Nur Çil
- Department of Molecular Medicine, Institute of Health Sciences, Dokuz Eylul University, Izmir, Turkey.
| | - Yasemin Soysal
- Department of Molecular Medicine, Institute of Health Sciences, Dokuz Eylul University, Izmir, Turkey
| |
Collapse
|
2
|
Diniz IMA, de Oliveira RF, do Valle IB, Picoli CC, Jácome-Santos H, de Almeida Queiroz Ferreira L, Avelar GF, Diniz MG, Birbrair A. Photobiomodulation therapy induces NG2 activation through dermal adipocyte lipolysis during wound healing. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2025; 267:113151. [PMID: 40187095 DOI: 10.1016/j.jphotobiol.2025.113151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/05/2025] [Accepted: 03/15/2025] [Indexed: 04/07/2025]
Abstract
Photobiomodulation therapy (PBMT) is a rapidly advancing approach for restoring damaged tissues, particularly in skin and mucosal wounds. While its application is promising, the role of mature adipocytes in regenerating mesenchymal tissues after PBMT remains largely unexplored. This study demonstrates that PBMT applied to skin wounds significantly reduces the number and size of mature adipocytes. Additionally, PBMT modulates the upregulation of peroxisome proliferator-activated receptor γ (PPARγ), increasing the gene expression of fatty acid binding protein 4 (Fabp4) and perilipin 1, which are linked to enhanced lipolysis. The molecular activation of neural/glial antigen 2 (NG2) indicates the recruitment of progenitor cells following mature adipocytes lipolysis. In vitro, PBMT improved dermal skin cell proliferation, migration, inflammatory regulation, and differentiation capacities. These findings reveal a novel mechanistic pathway for skin regeneration, emphasizing the therapeutic potential of PBMT in modulating dermal fat tissue to facilitate wound healing. Collectively, this emerging knowledge provides valuable insights into managing dermal fat tissue to support wound healing.
Collapse
Affiliation(s)
- Ivana Márcia Alves Diniz
- Department of Restorative Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| | - Rafaela Férrer de Oliveira
- Department of Restorative Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Isabella Bittencourt do Valle
- Department of Pathology, School of Dentistry, Universidade Federal do Espírito Santo, Vitória, Espírito Santo, Brazil
| | | | - Humberto Jácome-Santos
- Department of Restorative Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luiza de Almeida Queiroz Ferreira
- Department of Restorative Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Gleide Fernandes Avelar
- Department of Morphology, Biological Sciences Institute, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Marina Gonçalves Diniz
- Department of Pathology, Biological Sciences Institute, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Alexander Birbrair
- Department of Dermatology, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
3
|
Singh R, Katrolia A, Pal V. Chromenochalcones: a comprehensive review on developments towards a medicinal perspective. RSC Med Chem 2025:d5md00062a. [PMID: 40352673 PMCID: PMC12062893 DOI: 10.1039/d5md00062a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 04/18/2025] [Indexed: 05/14/2025] Open
Abstract
Chalcones are indeed a versatile scaffold in medicinal chemistry. Their structure, featuring an α,β-unsaturated carbonyl group, makes them highly reactive and capable of interacting with various biological targets. This reactivity is a key reason why chalcones and their derivatives are of such interest in drug discovery. The continued exploration of chalcone derivatives in medicinal chemistry will likely yield new insights and therapeutic candidates, given their broad spectrum of biological activities and the flexibility in modifying their structures. As chalcone derivatives, pyranochalcones and chromanchalcones are members of a subclass of flavonoids that are widely distributed. Several scientific databases were investigated to compile articles that illustrated the biological functions of chromenochalcones and their derivatives. Preclinical research on chromenochalcones and their derivatives is well covered in this review, highlighting the compounds with enormous significance as antimalarial, anti-inflammatory, antileishmanial, cytotoxic, antibacterial, antifungal, and antioxidant agents. In addition, the article briefly discusses the synthetic pathways employed for the total synthesis of selected pyranochalcones, including mallaophilippens C and E, citrunobin, and lesperol. Consequently, this overview may help research and design novel, potent therapeutic medications based on previously developed methodologies. This review is intended to provide a thorough, authoritative, and critical assessment of the chromenochalcone template for the chemistry community.
Collapse
Affiliation(s)
- Rohit Singh
- Department of Pharmaceutical Chemistry, College of Pharmacy, JSS University C-1/A, Sector-62 Noida Uttar Pradesh India
| | - Archita Katrolia
- Department of Pharmaceutics, College of Pharmacy, JSS University C-1/A, Sector-62 Noida Uttar Pradesh India
| | - Ved Pal
- Department of Pharmacognosy, College of Pharmacy, JSS University C-1/A, Sector-62 Noida Uttar Pradesh India
| |
Collapse
|
4
|
Gutiérrez-Hurtado IA, Martínez-López E, Rico-Méndez MA, Bravo-Villagra KM, Mendoza-Jaramillo HE, Sánchez-Rolón MDP, Betancourt-Núñez A, Gallegos-Arreola MP, Tapia-Rivera JC, López-Quintero A. Modulation of the Inflammatory Response by Adenovirus 36 in Patients with Obesity and Type 2 Diabetes: A Nested Case-Control Study Within a Cohort. Viruses 2025; 17:552. [PMID: 40284995 PMCID: PMC12030835 DOI: 10.3390/v17040552] [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: 02/21/2025] [Revised: 04/03/2025] [Accepted: 04/08/2025] [Indexed: 04/29/2025] Open
Abstract
Human adenovirus 36 (HAdV-36) is associated with obesity, potentially by promoting adipocyte proliferation and differentiation. Although linked to increased fat storage, HAdV-36 is also correlated with improved insulin sensitivity. Given its potential role in modulating adipose tissue and promoting a less inflammatory metabolic profile, its impacts on pro- and anti-inflammatory cytokine secretion remain unclear. METHODS This nested case-control study compared cytokine levels (IL-10, IL-2, IL-6, IL-8, and TNF-α) between patients with and without HAdV-36 infection. A total of 76 participants were included, with 37 in the control group (HAdV-36 negative) and 39 classified as cases (HAdV-36 positive). RESULTS HAdV-36 seropositive individuals exhibited significantly lower IL-6 levels and higher IL-8 levels than seronegative participants. Additionally, they had lower glucose levels, suggesting a potential link between HAdV-36 and metabolic regulation. CONCLUSIONS These findings support the hypothesis that HAdV-36 may influence inflammatory and metabolic responses by modulating cytokine expression and glucose levels. Further research is needed to clarify the underlying mechanisms and their implications for metabolic health.
Collapse
Affiliation(s)
- Itzae Adonai Gutiérrez-Hurtado
- Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (I.A.G.-H.); (E.M.-L.)
| | - Erika Martínez-López
- Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (I.A.G.-H.); (E.M.-L.)
- Instituto de Nutrigenética y Nutrigenómica Traslacional, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Manuel Alejandro Rico-Méndez
- Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (M.A.R.-M.); (K.M.B.-V.)
| | - Karla Mayela Bravo-Villagra
- Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (M.A.R.-M.); (K.M.B.-V.)
| | - Héctor Eduardo Mendoza-Jaramillo
- Departamento de Ciencias Básicas para la Salud, Centro Universitario del Sur, Universidad de Guadalajara, Ciudad Guzmán 49000, Mexico; (H.E.M.-J.); (M.d.P.S.-R.)
| | - María del Pilar Sánchez-Rolón
- Departamento de Ciencias Básicas para la Salud, Centro Universitario del Sur, Universidad de Guadalajara, Ciudad Guzmán 49000, Mexico; (H.E.M.-J.); (M.d.P.S.-R.)
| | - Alejandra Betancourt-Núñez
- Departamento de Disciplinas Filosófico, Metodológico e Instrumentales, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico;
| | - Martha Patricia Gallegos-Arreola
- División de Genética, Centro de Investigación Biomédica de Occidente (CIBO), Centro Médico Nacional de Occidente (CMNO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara 44340, Mexico;
| | - José Carlos Tapia-Rivera
- Departamento de Ciencias Básicas para la Salud, Centro Universitario del Sur, Universidad de Guadalajara, Ciudad Guzmán 49000, Mexico; (H.E.M.-J.); (M.d.P.S.-R.)
| | - Andres López-Quintero
- Instituto de Nutrigenética y Nutrigenómica Traslacional, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
- Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (M.A.R.-M.); (K.M.B.-V.)
| |
Collapse
|
5
|
Huo F, Liu C, Wang X, Li J, Wang Z, Liu D, Lan W, Zhu X, Lan J. SDCCAG3 inhibits adipocyte hypertrophy and improves obesity-related metabolic disorders via SDCCAG3/SMURF1/PPARγ axis. J Lipid Res 2025; 66:100772. [PMID: 40058593 PMCID: PMC12002885 DOI: 10.1016/j.jlr.2025.100772] [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: 01/03/2025] [Revised: 02/19/2025] [Accepted: 03/06/2025] [Indexed: 04/06/2025] Open
Abstract
Obesity is a prevalent global disease associated with various metabolic disorders. The expansion of white adipose tissue plays a pivotal role in regulating obesity-related metabolic dysfunctions. This study identified serum-defined colon cancer antigen 3 (SDCCAG3) as a novel key modulator of adipocyte metabolism. In adipose-specific SDCCAG3 knockout mice fed a high-fat diet, pathological expansion of adipose tissue, impaired glucose tolerance, insulin resistance, increased inflammatory markers, and augmented hepatic lipid accumulation were observed. Conversely, obesity models by specific overexpression of SDCCAG3 in adipose tissue confirmed that SDCCAG3 alleviated pathological expansion of adipose tissue, improved obesity-related metabolic disorders, with no observed changes in adipose tissue development under normal dietary conditions. Mechanistically, SDCCAG3 enhanced the stability of peroxisome proliferator-activated receptor gamma (PPARγ) by preventing its degradation via the ubiquitin-proteasome system through the SMAD specific E3 ubiquitin protein ligase 1 (SMURF1). Additionally, SDCCAG3 was subjected to negative transcriptional regulation by PPARγ, forming a SDCCAG3-PPARγ-SDCCAG3 loop that enhanced adipocyte lipid metabolism. Collectively, these findings demonstrated that SDCCAG3 functioned as a beneficial positive regulator of adipose tissue expansion and metabolic homeostasis, indicating its potential as a therapeutic target for metabolic diseases associated with nutrient excess.
Collapse
Affiliation(s)
- Fenglei Huo
- Department of Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Chenghang Liu
- Department of Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Xi Wang
- Department of Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Jinzheng Li
- College of Traditional Chinese Medicine, University of Traditional Chinese Medicine, Jinan, China
| | - Zhifeng Wang
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Duanqin Liu
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Weipeng Lan
- Department of Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Xingyan Zhu
- Department of Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Jing Lan
- Department of Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China.
| |
Collapse
|
6
|
Sawamoto A, Itagaki I, Okuyama S, Nakajima M. Reduction in MCP-1 production in preadipocytes is mediated by PPARγ activation and JNK/SIRT1 signaling. Biochim Biophys Acta Gen Subj 2025; 1869:130737. [PMID: 39672476 DOI: 10.1016/j.bbagen.2024.130737] [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: 09/23/2024] [Revised: 12/03/2024] [Accepted: 12/08/2024] [Indexed: 12/15/2024]
Abstract
Obesity-induced monocyte chemoattractant protein 1 (MCP-1) production leads to the infiltration of monocytes/macrophages into white adipose tissue (WAT), which contributes to systemic insulin resistance. Peroxisome proliferator-activated receptor gamma (PPARγ) agonists are known to reduce MCP-1 production in both humans and mice; however, the underlying mechanism in WAT remains unclear. Here, we propose a novel mechanism for the reduction in MCP-1 production in preadipocytes. The PPARγ agonist rosiglitazone (RSG) reduced MCP-1 production and secretion in response to lipopolysaccharide (LPS) in 3T3-L1 preadipocytes and mouse stromal vascular fraction-derived primary preadipocytes. Both RSG and SP600125 (a c-Jun N-terminal kinase (JNK) inhibitor) inhibited LPS-induced degradation of silent information regulator 2 homolog 1 (SIRT1), a negative regulator of MCP-1 production in 3T3-L1 preadipocytes. Furthermore, RSG inhibited LPS-induced activation of nuclear factor-κB. These effects of RSG were abolished in 3T3-L1 preadipocytes transfected with Pparg siRNA. These findings highlight a novel mechanism by which PPARγ activation inhibits JNK/SIRT1 signaling in preadipocytes and contributes to the reduction in MCP-1 production, suggesting that preadipocytes could be a potential therapeutic target for the treatment of insulin resistance.
Collapse
Affiliation(s)
- Atsushi Sawamoto
- Department of Pharmaceutical Pharmacology, College of Pharmaceutical Sciences, Matsuyama University, 4-2 Bunkyo-cho, Matsuyama, Ehime 790-8578, Japan.
| | - Ibuki Itagaki
- Department of Pharmaceutical Pharmacology, College of Pharmaceutical Sciences, Matsuyama University, 4-2 Bunkyo-cho, Matsuyama, Ehime 790-8578, Japan
| | - Satoshi Okuyama
- Department of Pharmaceutical Pharmacology, College of Pharmaceutical Sciences, Matsuyama University, 4-2 Bunkyo-cho, Matsuyama, Ehime 790-8578, Japan
| | - Mitsunari Nakajima
- Department of Pharmaceutical Pharmacology, College of Pharmaceutical Sciences, Matsuyama University, 4-2 Bunkyo-cho, Matsuyama, Ehime 790-8578, Japan
| |
Collapse
|
7
|
Chen F, Ma L, Liu Q, Zhou Z, Yi W. Recent advances and therapeutic applications of PPARγ-targeted ligands based on the inhibition mechanism of Ser273 phosphorylation. Metabolism 2025; 163:156097. [PMID: 39637972 DOI: 10.1016/j.metabol.2024.156097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/27/2024] [Accepted: 11/30/2024] [Indexed: 12/07/2024]
Abstract
PPARγ functions as a master ligand-dependent transcription factor that regulates the expressions of a variety of key genes related to metabolic homeostasis and inflammatory immunity. It has been recognized as a popular and druggable target in modern drug discovery. Similar to other nuclear receptors, PPARγ is a phosphoprotein, and its biological functions are regulated by phosphorylation, especially at Ser273 site which is mediated by CDK5 or ERK. In the past decade, the excessive level of PPARγ-Ser273 phosphorylation has been confirmed to be a crucial factor in promoting the occurrence and development of some major diseases. Ligands capable of inhibiting PPARγ-Ser273 phosphorylation have shown great potentials for treatment. Despite these achievements, to our knowledge, no related review focusing on this topic has been conducted so far. Therefore, we herein summarize the basic knowledge of PPARγ and CDK5/ERK-mediated PPARγ-Ser273 phosphorylation as well as its physiopathological role in representative diseases. We also review the developments and therapeutic applications of PPARγ-targeted ligands based on this mechanism. Finally, we suggest several directions for future investigations. We expect that this review can evoke more inspiration of scientific communities, ultimately facilitating the promotion of the PPARγ-Ser273 phosphorylation-involved mechanism as a promising breakthrough point for addressing the clinical treatment of human diseases.
Collapse
Affiliation(s)
- Fangyuan Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation & Molecular Target and Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Lei Ma
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation & Molecular Target and Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Qingmei Liu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation & Molecular Target and Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Zhi Zhou
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation & Molecular Target and Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China.
| | - Wei Yi
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation & Molecular Target and Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China.
| |
Collapse
|
8
|
Miyata R, Suzuki M, Okazaki Y, Abe D, Nakajima Y. Peroxisome Proliferator-Activated Receptor-Gamma Activation by an Active Compound in Lythrum anceps (Koehne) Makino. J Cell Biochem 2025; 126:e70009. [PMID: 39980354 DOI: 10.1002/jcb.70009] [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: 12/06/2024] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/22/2025]
Abstract
Lythrum anceps (Koehne) Makino (Japanese common name: "Misohagi") is an edible plant belonging to the Lythraceae family. It is mainly distributed in Asia, Northern Africa, and Europe. Plants of the genus Lythrum exhibit a broad range of biological activities including anti-inflammatory and antimicrobial activities. Because of this, the plants are used in traditional medicine to treat hemorrhage, infected wounds, and dysentery. The activation of peroxisome proliferator-activated receptor-gamma (PPARγ) is an effective target for improving insulin resistance and anti-inflammatory activity. However, PPARγ activation by the genus Lythrum remains unclear. Aiming to evaluate PPARγ activation by L. anceps, we generated a reporter cell line using an artificial chromosome vector that stably expresses dual-color beetle luciferases. Dual-color real-time bioluminescence monitoring revealed marked PPARγ activation in L. anceps extracts. Moreover, ellagic acid was identified as a PPARγ activator present in L. anceps by a bioassay-guided fractionation approach.
Collapse
Affiliation(s)
- Ryo Miyata
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Takamatsu, Kagawa, Japan
| | - Masanobu Suzuki
- Resources and Environment Division, Kochi Prefectural Industrial Technology Center, Kochi, Kochi, Japan
| | - Yuka Okazaki
- Resources and Environment Division, Kochi Prefectural Industrial Technology Center, Kochi, Kochi, Japan
| | - Daigo Abe
- Western Region Agricultural Research Center, National Agricultural and Food Research Organization (NARO), Zentsuji, Kagawa, Japan
| | - Yoshihiro Nakajima
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Takamatsu, Kagawa, Japan
| |
Collapse
|
9
|
Hayes CM, Gallucci GM, Boyer JL, Assis DN, Ghonem NS. PPAR agonists for the treatment of cholestatic liver diseases: Over a decade of clinical progress. Hepatol Commun 2025; 9:e0612. [PMID: 39699308 PMCID: PMC11661771 DOI: 10.1097/hc9.0000000000000612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 11/13/2024] [Indexed: 12/20/2024] Open
Abstract
Primary biliary cholangitis (PBC) and primary sclerosing cholangitis (PSC) are characterized by the destruction of the small bile ducts and the formation of multifocal biliary strictures, respectively, impairing bile flow. This leads to the hepatic accumulation of bile acids, causing liver injury and the risk of progression to cirrhosis and liver failure. First-line therapy for PBC is ursodeoxycholic acid, although up to 40% of treated individuals are incomplete responders, and there is no effective therapy for PSC, highlighting the need for better therapeutic options in these diseases. In addition, pruritus is a common symptom of cholestasis that has severe consequences for quality of life and is often undertreated or untreated. Nuclear receptors are pharmacological targets to treat cholestasis due to their multifactorial regulation of hepatic enzymatic pathways, particularly in bile acid metabolism. The peroxisome proliferator-activated receptor (PPAR) is of significant clinical interest due to its role in regulating bile acid synthesis and detoxification pathways. PPAR agonism by fibrates has traditionally been explored due to PPARα's expression in the liver; however, recent interest has expanded to focus on newer PPAR agonists that activate other PPAR isoforms, for example, δ, γ, alone or in combination. Several PPAR agonists have been investigated as second-line therapy for people living with PBC, including the recent accelerated United States Food and Drug Administration approval of elafibranor and seladelpar. This review evaluates available data on the efficacy and safety of the five PPAR agonists investigated for the treatment of cholestasis and associated pruritus in PBC and PSC, namely fenofibrate, bezafibrate, saroglitazar, elafibranor, and seladelpar.
Collapse
Affiliation(s)
- Colleen M. Hayes
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
| | - Gina M. Gallucci
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
| | - James L. Boyer
- Section of Digestive Diseases and Yale Liver Center, Yale School of Medicine, New Haven, Connecticut, USA
| | - David N. Assis
- Section of Digestive Diseases and Yale Liver Center, Yale School of Medicine, New Haven, Connecticut, USA
| | - Nisanne S. Ghonem
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
| |
Collapse
|
10
|
Özdaş S, Canatar İ, Derici GE, Koç M. Bolanthus turcicus: a promising antidiabetic with in-vitro antioxidant, enzyme inhibitory and antiadipogenic activities. J Mol Histol 2024; 56:59. [PMID: 39729235 DOI: 10.1007/s10735-024-10283-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/01/2024] [Indexed: 12/28/2024]
Abstract
It is crucial to investigate new anti-diabetic agents and therapeutic approaches targeting molecules in potential signaling pathways for the treatment of Type 2 diabetes mellitus (T2DM). The objective of the study was to investigate the total phenolic content, antioxidant capacity, α-glucosidase, and α-amylase inhibitory activities of Bolanthus turcicus (B. turcicus), as well as their cytotoxic, anti-adipogenic, anti-diabetic, apoptotic, and anti-migration potential on adipocytes. B. turcicus samples were extracted with methanol (MeOH), ethyl acetate (EA) and aqueous (Aq) solvents. The MeOH extract had the highest phenolic content (81.14 mg GAE/g), followed by EA (74.93 mg GAE/g) and Aq (51.09 mg GAE/g). All extracts exhibited dose-dependent increases in α-glycosidase and α-amylase inhibitory activity. B. turcicus extracts showed cytotoxic effect on adipocytes with IC50 values of MeOH (141.0 µg/mL) < Aq (155.3 µg/mL) < EA (199.5 µg/mL). Furthermore, B. turcicus extracts reduced lipid droplet formation and adipocyte diameter size. All extracts altered cell morphology to resemble fibroblasts. B. turcicus extracts exhibited anti-migratory effect delaying wound healing for up to 96 h. The B. turcicus extracts showed a pro-apoptotic effects on adipocytes by increasing Caspase-3 enzyme activity and the population of DAPI-positive cell with apoptotic nuclear-morphology. B. turcicus extracts upregulated the expression of the Glut-4 gene at the mRNA, protein and intracellular level in adipocytes. In conclusion, our findings indicate that B. turcicus not only exhibits strong antioxidant properties and enzyme inhibitory activities but also exerts significant anti-adipogenic and pro-apoptotic effects in adipocytes, thereby providing a comprehensive mechanism through which it may contribute to the management of T2DM. These effects highlight the potential of B. turcicus as a therapeutic agent for improving glucose homeostasis and insulin sensitivity.
Collapse
Affiliation(s)
- Sibel Özdaş
- Department of Bioengineering, Adana Alparslan Türkeş Science and Technology University, Adana, Türkiye, Turkey.
| | - İpek Canatar
- Department of Bioengineering, Adana Alparslan Türkeş Science and Technology University, Adana, Türkiye, Turkey
| | - Gizem Ece Derici
- Department of Bioengineering, Adana Alparslan Türkeş Science and Technology University, Adana, Türkiye, Turkey
| | - Murat Koç
- Complementary and Integrative Medicine, Department of Traditional, Ankara Yıldırım Beyazıt University, Ankara, Türkiye, Turkey
| |
Collapse
|
11
|
Bahadoran Z, Mirmiran P, Kashfi K, Ghasemi A. Effects of time-restricted feeding (TRF)-model of intermittent fasting on adipose organ: a narrative review. Eat Weight Disord 2024; 29:77. [PMID: 39719521 DOI: 10.1007/s40519-024-01709-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 12/11/2024] [Indexed: 12/26/2024] Open
Abstract
Time-restricted feeding (TRF), an intermittent fasting approach involving a shortened eating window within 24 h, has gained popularity as a weight management approach. This review addresses how TRF may favor fat redistribution and the function of the adipose organ. TRF trials (mainly 16:8 model, with a duration of 5-48 weeks) reported a significant weight loss (1.2-10.2%, ~ 1.4-9.4 kg), with a considerable decrease in total fat mass (1.6-21%, ~ 0.5-7 kg) and visceral adipose compartment (VAC, 11-27%) in overweight and obese subjects. Experimental TRF in normal-fed and obesogenic-diet-fed mice and rats (with a fasting duration ranging between 9 and 21 h within 1-17 weeks) reported a significant reduction in body weight (~ 7-40%), total fat mass (~ 17-71%), and intrahepatic fat (~ 25-72%). TRF also improves VAC and subcutaneous adipose compartment (SAC) function by decreasing adipocyte size, macrophage infiltration, M1-macrophage polarity, and downregulating inflammatory genes. In conclusion, beyond its effect on body weight loss, total fat mass, and intrahepatic fat accumulation, TRF favors adipose organ fat redistribution in overweight and obese subjects by decreasing VAC and improving the function of VAC and SAC.
Collapse
Affiliation(s)
- Zahra Bahadoran
- Micronutrient Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, 10031, USA
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, No. 24, Sahid-Erabi St, Yemen St, Chamran Exp, Tehran, Iran.
| |
Collapse
|
12
|
Deng S, Zhang S, Shen T, Wang X, Gao Z, Zhang W, Dai K, Wang J, Liu C. Amphiphilic cytokine traps remodel marrow adipose tissue for hematopoietic microenvironment amelioration. Bioact Mater 2024; 42:226-240. [PMID: 39285915 PMCID: PMC11404087 DOI: 10.1016/j.bioactmat.2024.08.032] [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: 03/25/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is extensively employed in the treatment of hematological malignancies but is markedly constrained by the paucity of hematopoietic stem/progenitor cells (HSPCs). Recent studies have found that marrow adipose tissue (MAT) acts on hematopoiesis through complicated mechanisms. Therefore, the osteo-organoids fabricated in vivo using biomaterials loaded with recombinant human bone morphogenetic protein 2 (rhBMP-2) have been used as models of MAT for our research. To obtain sufficient amounts of therapeutic HSPCs and healthy MAT, we have developed amphiphilic chitosan (AC)-gelatin as carriers of rhBMP-2 to the regulate type conversion of adipose tissue and trap hematopoietic growth factors. Unlike medicine interventions or cell therapies, the traps based on AC not only attenuate the occupancy of adipocytes within the hematopoietic microenvironment while preserving stem cell factor concentrations, but also improve marrow metabolism by promoting MAT browning. In conclusion, this approach increases the proportion of HSPCs in osteo-organoids, and optimizes the composition and metabolic status of MAT. These findings furnish an experimental basis for regulating hematopoiesis in vivo through materials that promote the development of autologous HSPCs. Additionally, this approach presents a theoretical model of rapid adipogenesis for the study of adipose-related pathologies and potential pharmacological targets.
Collapse
Affiliation(s)
- Shunshu Deng
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092, PR China
- Key Laboratory for Ultrafine Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Shuang Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Key Laboratory for Ultrafine Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Tong Shen
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Key Laboratory for Ultrafine Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Xuanlin Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Key Laboratory for Ultrafine Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Zehua Gao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Key Laboratory for Ultrafine Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Wenchao Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Key Laboratory for Ultrafine Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Kai Dai
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Key Laboratory for Ultrafine Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Jing Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, PR China
| |
Collapse
|
13
|
Chen X, Mo X, Zhang Y, He D, Xiao R, Cheng Q, Wang H, Liu L, Li WW, Xie P. A comprehensive analysis of the differential expression in the hippocampus of depression induced by gut microbiota compared to traditional stress. Gene 2024; 927:148633. [PMID: 38838871 DOI: 10.1016/j.gene.2024.148633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 05/22/2024] [Accepted: 05/30/2024] [Indexed: 06/07/2024]
Abstract
Depression, which is a disease of heterogeneous etiology, is characterized by high disability and mortality rates. Gut microbiota are associated with the development of depression. To further explore any differences in the mechanisms of depression induced by gut microbiota and traditional stresses, as well as facilitate the development of microbiota-based interventions, a fecal microbiota transplantation (FMT) depression model was made. This was achieved by transplanting feces from major depressive disorder (MDD) patients into germ-free mice. Second, the mechanisms of the depression induced by gut microbiota were analyzed in comparison with those of the depression caused by different forms of stress. It turned out that mice exhibited depressive-like behavior after FMT. Then, PCR array analysis was performed on the hippocampus of the depressed mice to identify differentially expressed genes (DEGs). The KEGG analysis revealed that the pathways of depression induced by gut microbes are closely associated with immuno-inflammation. To determine the pathogenic pathways of physiological stress and psychological stress-induced depression, raw data was extracted from several databases and KEGG analysis was performed. The results from the analysis revealed that the mechanisms of depression induced by physiological and psychological stress are closely related to the regulation of neurotransmitters and energy metabolism. Interestingly, the immunoinflammatory response was distinct across different etiologies that induced depression. The findings showed that gut microbiota dysbiosis-induced depression was mainly associated with adaptive immunity, while physiological stress-induced depression was more linked to innate immunity. This study compared the pathogenesis of depression caused by gut microbiota dysbiosis, and physiological and psychological stress. We explored new intervention methods for depression and laid the foundation for precise treatment.
Collapse
Affiliation(s)
- Xueyi Chen
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; School of Basic Medical Sciences, Department of Pathology, Chongqing Medical University, Chongqing 400016, China
| | - Xiaolong Mo
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yangdong Zhang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Dian He
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Rui Xiao
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; School of Basic Medical Sciences, Department of Pathology, Chongqing Medical University, Chongqing 400016, China
| | - Qisheng Cheng
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Haiyang Wang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Lanxiang Liu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing 402160, China
| | - Wen-Wen Li
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; School of Basic Medical Sciences, Department of Pathology, Chongqing Medical University, Chongqing 400016, China; Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing 400016, China; Department of Pathology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Peng Xie
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing 402160, China.
| |
Collapse
|
14
|
Sun L. Associations between waist-to-height ratio and abdominal aortic calcification: A cross-sectional study. Medicine (Baltimore) 2024; 103:e38608. [PMID: 38875360 PMCID: PMC11175898 DOI: 10.1097/md.0000000000038608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/16/2024] Open
Abstract
Waist-to-height ratio (WtHR) is a validated biomarker of central obesity that appears to be preferable to other body composition measurements in the evaluation of cardiovascular disease. The goal of this research was to explore the connection between WtHR and abdominal aortic calcification (AAC) among adults. On the basis of data from the 2013 to 2014 National Health and Nutrition Examination Survey, multivariate logistic regression, sensitivity analysis, as well as smoothed curve fitting were used to evaluate the connection between WtHR and AAC. Subgroup analyses along with interaction tests were done to see if this link was consistent across populations. Among 3079 participants aged >40 years, there was a negative association between WtHR and ACC. Each 1-unit emergence of WtHR was related to a 2% reduction in the probability of severe AAC in the entirely adjusted model (odds ratio = 0.02, 95% confidence interval: [0.00-0.12]). Participants in the highest WtHR quartile were 39% less likely to acquire severe AAC compared with those in the lowest quartile. (odds ratio = 0.61, 95% confidence interval: [0.37-1.00]). This negative association was more pronounced in the diabetes subgroup. We discovered a reversed U-shaped association between WtHR as well as AAC score utilizing a 2-stage linear regression model, with an intersection point of 0.56. WtHR was negatively associated with AAC among US adults.
Collapse
Affiliation(s)
- Lijie Sun
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
| |
Collapse
|
15
|
Wang Y, Lei F, Lin Y, Han Y, Yang L, Tan H. Peroxisome proliferator-activated receptors as therapeutic target for cancer. J Cell Mol Med 2024; 28:e17931. [PMID: 37700501 PMCID: PMC10902584 DOI: 10.1111/jcmm.17931] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/05/2023] [Accepted: 08/18/2023] [Indexed: 09/14/2023] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are transcription factors belonging to the nuclear receptor family. There are three subtypes of PPARs, including PPAR-α, PPAR-β/δ and PPAR-γ. They are expressed in different tissues and act by regulating the expression of target genes in the form of binding to ligands. Various subtypes of PPAR have been shown to have significant roles in a wide range of biological processes including lipid metabolism, body energy homeostasis, cell proliferation and differentiation, bone formation, tissue repair and remodelling. Recent studies have found that PPARs are closely related to tumours. They are involved in cancer cell growth, angiogenesis and tumour immune response, and are essential components in tumour progression and metastasis. As such, they have become a target for cancer therapy research. In this review, we discussed the current state of knowledge on the involvement of PPARs in cancer, including their role in tumourigenesis, the impact of PPARs in tumour microenvironment and the potential of using PPARs combinational therapy to treat cancer by targeting essential signal pathways, or as adjuvants to boost the effects of current chemo and immunotherapies. Our review highlights the complexity of PPARs in cancer and the need for a better understanding of the mechanism in order to design effective cancer therapies.
Collapse
Affiliation(s)
- Yuqing Wang
- Department of Internal MedicineMontefiore Medical Center, Wakefield CampusBronxNew YorkUSA
| | - Feifei Lei
- Department of Infectious Disease, Lab of Liver Disease, Renmin HospitalHubei University of MedicineShiyanChina
| | - Yiyun Lin
- Department of Biomedical SciencesUniversity of Texas, MD Anderson Cancer CenterHoustonTexasUSA
| | - Yuru Han
- Qinghai Provincial People's HospitalXiningChina
| | - Lei Yang
- Department of Biomedical SciencesUniversity of Texas, MD Anderson Cancer CenterHoustonTexasUSA
| | - Huabing Tan
- Department of Infectious Disease, Lab of Liver Disease, Renmin HospitalHubei University of MedicineShiyanChina
| |
Collapse
|
16
|
Zhang F, Pan X, Zhang X, Tong N. The effect of thiazolidinediones on body fat redistribution in adults: A systematic review and meta-analysis of randomized controlled trials. Obes Rev 2024; 25:e13675. [PMID: 38098209 DOI: 10.1111/obr.13675] [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: 08/29/2022] [Revised: 11/04/2023] [Accepted: 11/10/2023] [Indexed: 02/28/2024]
Abstract
Visceral adiposity is a strong predictor of cardiometabolic risk. Thiazolidinediones (TZDs) are associated with a shift in fat redistribution from visceral adipose tissue (VAT) to subcutaneous adipose tissue (SAT). We aimed to compare the effects of TZD and other interventions on fat remodeling in adults in randomized controlled trials. Among the 1331 retrieved studies, 39 trials with 1765 participants were included in the meta-analysis. The standardized mean difference in VAT change was not significantly different between TZD and comparators across the overall studies. Intriguingly, TZD treatment resulted in significant decreases in VAT compared with placebo and sulfonylureas (p < 0.05), although recombinant human growth hormone was superior to TZD regarding VAT reduction (p < 0.05). Data from 216 participants showed TZD leading to a greater reduction in liver fat percentage than comparators (p < 0.05). Compared with the controls, TZD significantly increased SAT, total body fat, weight, waist circumference, and body mass index (p < 0.05). However, TZD pronouncedly improved glucose control, insulin resistance, adiponectin, and lipid profile (p < 0.05). TZD provides a favorable effect on fat redistribution and benefits insulin sensitivity, suggesting a potentially valuable approach in cardiometabolic risk management.
Collapse
Affiliation(s)
- Fang Zhang
- Division of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xiaohui Pan
- Division of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xinyue Zhang
- Division of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Nanwei Tong
- Division of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
17
|
Safdar M, Hassan F, Khan MS, Khan AH, Junejo Y, Ozaslan M, Arain MA, Behan AA. In silico analysis of polyphenols modulate bovine PPARγ to increase milk fat synthesis in dairy cattle via the MAPK signaling pathways. J Anim Sci 2024; 102:skae248. [PMID: 39210246 PMCID: PMC11551727 DOI: 10.1093/jas/skae248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
This study investigates the potential phytochemicals that modulate bovine peroxisome proliferator-activated receptor gamma (PPARγ) and the mitogen-activated protein kinase (MAPK) pathways to enhance milk fat production in dairy animals. Bovine PPARγ, a key member of the nuclear hormone receptor superfamily, plays a vital role in regulating metabolic, cellular differentiation, apoptosis, and anti-inflammatory responses in livestock, while the MAPK pathway is contributory in cellular processes that impact milk fat synthesis. This approach involved an all-inclusive molecular docking analysis of 10,000 polyphenols to identify potential PPARγ ligands. From this extensive screening, top 10 compounds were selected that exhibited the highest binding affinities to bovine PPARγ. Particularly, curcumin sulfate, isoflavone, and quercetin emerged as the most promising candidates. These compounds demonstrated superior docking scores (-9.28 kcal/mol, -9.27 kcal/mol, and -7.31 kcal/mol, respectively) and lower RMSD values compared to the synthetic bovine PPARγ agonist, 2,4-thiazolidinedione (-4.12 kcal/mol), indicating a strong potential for modulating the receptor. Molecular dynamics simulations (MDS) further affirmed the stability of these polyphenols-bovine PPARγ complexes, suggesting their effective and sustained interactions. These polyphenols, known as fatty acid synthase inhibitors, are suggested to influence lipid metabolism pathways crucial to milk fat production, possibly through the downregulation of the MAPK pathway. The screened compounds showed favorable pharmacokinetic profiles, including nontoxicity, carcinogenicity, and high gastrointestinal absorption, positioning them as viable candidates for enhancing dairy cattle health and milk production. These findings may open new possibilities for the use of phytochemicals as feed additives in dairy animals, suggesting a novel approach to improve milk fat synthesis through the dual modulation of bovine PPARγ and MAPK pathways.
Collapse
Affiliation(s)
- Muhammad Safdar
- Faculty of Animal Production and Technology, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
| | - Faizul Hassan
- Faculty of Animal Production and Technology, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
| | - Muhammad Sajjad Khan
- Faculty of Animal Production and Technology, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
| | - Aneeb Hassan Khan
- Faculty of Animal Production and Technology, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
| | - Yasmeen Junejo
- Faculty of Animal Production and Technology, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
| | - Mehmet Ozaslan
- Department of Biology, Division of Molecular Biology and Genetics, Gaziantep University, Gaziantep, Turkey
| | - Muhammad Asif Arain
- Faculty of Veterinary and Animal Sciences, Lasbela University of Agriculture, Water and Marine Sciences, Uthal, Pakistan
| | - Atique Ahmed Behan
- Department of Animal and Veterinary Sciences, College of Agricultural and Marine Sciences, Sultan Qaboos University, Muscat, Oman
| |
Collapse
|
18
|
Azizogli AR, Vitti MR, Mishra R, Osorno L, Heffernan C, Kumar VA. Comparison of SGLT1, SGLT2, and Dual Inhibitor biological activity in treating Type 2 Diabetes Mellitus. ADVANCED THERAPEUTICS 2023; 6:2300143. [PMID: 38223846 PMCID: PMC10783160 DOI: 10.1002/adtp.202300143] [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: 04/25/2023] [Indexed: 01/16/2024]
Abstract
Diabetes Mellitus Type 2 (T2D) is an emerging health burden in the USand worldwide, impacting approximately 15% of Americans. Current front-line therapeutics for T2D patients include sulfonylureas that act to reduce A1C and/or fasting blood glucose levels, or Metformin that antagonizes the action of glucagon to reduce hepatic glucose production. Next generation glucomodulatory therapeutics target members of the high-affinity glucose transporter Sodium-Glucose-Linked-Transporter (SGLT) family. SGLT1 is primarily expressed in intestinal epithelium, whose inhibition reduces dietary glucose uptake, whilst SGLT2 is highly expressed in kidney - regulating glucose reabsorption. A number of SGLT2 inhibitors are FDA approved whilst SGLT1 and dual SGLT1 & 2 inhibitor are currently in clinical trials. Here, we discuss and compare SGLT2, SGLT1, and dual inhibitors' biochemical mechanism and physiological effects.
Collapse
Affiliation(s)
- Abdul-Rahman Azizogli
- Department of Biological Sciences, New Jersey Institute of Technology, Newark, NJ, 07102
| | - Michael R Vitti
- University of Virginia School of Medicine, Charlottesville, VA, 22903
| | - Richa Mishra
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102
| | - Laura Osorno
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102
| | - Corey Heffernan
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102
| | - Vivek A Kumar
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102
- Department of Biological Sciences, New Jersey Institute of Technology, Newark, NJ, 07102
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, 07102
- Department of Endodontics, Rutgers School of Dental Medicine, Newark, NJ, 07103
| |
Collapse
|
19
|
Kasahara N, Imi Y, Amano R, Shinohara M, Okada K, Hosokawa Y, Imamori M, Tomimoto C, Kunisawa J, Kishino S, Ogawa J, Ogawa W, Hosooka T. A gut microbial metabolite of linoleic acid ameliorates liver fibrosis by inhibiting TGF-β signaling in hepatic stellate cells. Sci Rep 2023; 13:18983. [PMID: 37923895 PMCID: PMC10624680 DOI: 10.1038/s41598-023-46404-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 10/31/2023] [Indexed: 11/06/2023] Open
Abstract
The antidiabetic drug pioglitazone ameliorates insulin resistance by activating the transcription factor PPARγ. In addition to its blood glucose-lowering action, pioglitazone exerts pleiotropic effects including amelioration of nonalcoholic fatty liver disease (NAFLD)/nonalcoholic steatohepatitis (NASH). The mechanism by which pioglitazone achieves this latter effect has remained unclear, however. We here show that pioglitazone administration increases the amount of linoleic acid (LA) metabolites in adipose tissue of KK-Ay mice. These metabolites are produced by lactic acid bacteria in the gut, and pioglitazone also increased the fraction of Lactobacillus in the gut microbiota. Administration of the LA metabolite HYA (10-hydroxy-cis-12-octadecenoic acid) to C57BL/6 J mice fed a high-fat diet improved liver histology including steatosis, inflammatory cell infiltration, and fibrosis. Gene ontology analysis of RNA-sequencing data for the liver revealed that the top category for genes downregulated by HYA treatment was related to extracellular matrix, and the expression of individual genes related to fibrosis was confirmed to be attenuated by HYA treatment. Mechanistically, HYA suppressed TGF-β-induced Smad3 phosphorylation and fibrosis-related gene expression in human hepatic stellate cells (LX-2). Our results implicate LA metabolites in the mechanism by which pioglitazone ameliorates liver fibrosis, and they suggest that HYA is a potential therapeutic for NAFLD/NASH.
Collapse
Affiliation(s)
- Nanaho Kasahara
- Laboratory of Nutritional Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-Ku, Shizuoka, 422-8526, Japan
| | - Yukiko Imi
- Laboratory of Nutritional Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-Ku, Shizuoka, 422-8526, Japan
| | - Reina Amano
- Laboratory of Nutritional Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-Ku, Shizuoka, 422-8526, Japan
| | - Masakazu Shinohara
- Division of Molecular Epidemiology, Department of Future Medicine Sciences, Kobe University Graduate School of Medicine, Kobe, Hyogo, 650-0017, Japan
- The Integrated Center for Mass Spectrometry, Kobe University Graduate School of Medicine, Kobe, Hyogo, 650-0017, Japan
| | - Kumiko Okada
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo, 650-0017, Japan
| | - Yusei Hosokawa
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo, 650-0017, Japan
| | - Makoto Imamori
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo, 650-0017, Japan
| | | | - Jun Kunisawa
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health, and Nutrition (NIBIOHN), Osaka, 567-0085, Japan
| | - Shigenobu Kishino
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo-Ku, Kyoto, 606-8502, Japan
| | - Jun Ogawa
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo-Ku, Kyoto, 606-8502, Japan
| | - Wataru Ogawa
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo, 650-0017, Japan
| | - Tetsuya Hosooka
- Laboratory of Nutritional Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-Ku, Shizuoka, 422-8526, Japan.
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo, 650-0017, Japan.
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health, and Nutrition (NIBIOHN), Osaka, 567-0085, Japan.
| |
Collapse
|
20
|
Bajetto A, Pattarozzi A, Sirito R, Barbieri F, Florio T. Metformin potentiates immunosuppressant activity and adipogenic differentiation of human umbilical cord-mesenchymal stem cells. Int Immunopharmacol 2023; 124:111078. [PMID: 37844465 DOI: 10.1016/j.intimp.2023.111078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/25/2023] [Accepted: 10/11/2023] [Indexed: 10/18/2023]
Abstract
Metformin, a first-line drug for type-2 diabetes, displays pleiotropic effects on inflammation, aging, and cancer. Obesity triggers a low-grade chronic inflammation leading to insulin resistance, characterized by increased pro-inflammatory cytokines produced by adipocytes and infiltrated immune cells, which contributes to metabolic syndrome. We investigated metformin's differentiation and immunoregulatory properties of human umbilical cord-mesenchymal stem cells (UC-MSC), as cellular basis of its beneficial role in metabolic dysfunctions. Isolation, characterization and multilineage differentiation of UC-MSC were performed using standard protocols and flow-cytometry. Metformin effects on UC-MSC growth was assessed by colony formation and MTT assay, gene and protein expression by qRT-PCR, and western blot analysis. Proliferation of peripheral blood mononuclear cells (PBMCs) co-cultured with metformin-treated UC-MSC-conditioned media was evaluated by dye dilution assay. We show that metformin decreases proliferation and colony formation of UC-MSCs and enhances their adipogenic lineage commitment. Metformin (3 mM) increases PPARγ and downregulates FABP4 mRNA both in basal and in adipogenic culture conditions; however, the modulation of PPARγ expression is unrelated to the antiproliferative effects. Moreover, metformin inhibits UC-MSC inflammatory activity reducing the expression of IL-6, MCP-1, and COX-2. Conditioned media, collected from metformin-treated UC-MSCs, down-regulate CD3+ T lymphocyte growth in stimulated PBMCs and, in particular, reduce the CD8+ T cell population. These results indicate that metformin may favor new adipocyte formation and potentiate immune suppressive properties of UC-MSCs. Thus, adipose tissue regeneration and anti-inflammatory activity may represent possible mechanisms by which metformin exerts its positive effect on lipid metabolism.
Collapse
Affiliation(s)
- Adriana Bajetto
- Section of Pharmacology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy
| | - Alessandra Pattarozzi
- Section of Pharmacology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy
| | - Rodolfo Sirito
- Section of Obstetrics and Gynaecology, International Evangelical Hospital, 16122 Genova, Italy
| | - Federica Barbieri
- Section of Pharmacology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy
| | - Tullio Florio
- Section of Pharmacology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy; IRCCS, Ospedale Policlinico San Martino, 16132 Genova, Italy.
| |
Collapse
|
21
|
Palacios-Marin I, Serra D, Jimenez-Chillarón J, Herrero L, Todorčević M. Adipose Tissue Dynamics: Cellular and Lipid Turnover in Health and Disease. Nutrients 2023; 15:3968. [PMID: 37764752 PMCID: PMC10535304 DOI: 10.3390/nu15183968] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 08/30/2023] [Accepted: 09/10/2023] [Indexed: 09/29/2023] Open
Abstract
The alarming increase in obesity and its related metabolic health complications, such as type 2 diabetes, has evolved into a global pandemic. Obesity is mainly characterized by excessive accumulation of adipose tissue, primarily due to an imbalance between energy intake and expenditure. Prolonged positive energy balance leads to the expansion of existing adipocytes (hypertrophy) and/or an increase in preadipocyte and adipocyte number (hyperplasia) to accommodate excess energy intake. However, obesity is not solely defined by increases in adipocyte size and number. The turnover of adipose tissue cells also plays a crucial role in the development and progression of obesity. Cell turnover encompasses the processes of cell proliferation, differentiation, and apoptosis, which collectively regulate the overall cell population within adipose tissue. Lipid turnover represents another critical factor that influences how adipose tissue stores and releases energy. Our understanding of adipose tissue lipid turnover in humans remains limited due to the slow rate of turnover and methodological constraints. Nonetheless, disturbances in lipid metabolism are strongly associated with altered adipose tissue lipid turnover. In obesity, there is a decreased rate of triglyceride removal (lipolysis followed by oxidation), leading to the accumulation of triglycerides over time. This review provides a comprehensive summary of findings from both in vitro and in vivo methods used to study the turnover of adipose cells and lipids in metabolic health and disease. Understanding the mechanisms underlying cellular and lipid turnover in obesity is essential for developing strategies to mitigate the adverse effects of excess adiposity.
Collapse
Affiliation(s)
- Ivonne Palacios-Marin
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, Esplugues, E-08950 Barcelona, Spain
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain
| | - Dolors Serra
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Josep Jimenez-Chillarón
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, Esplugues, E-08950 Barcelona, Spain
- Department of Physiological Sciences, School of Medicine, University of Barcelona, E-08907 L’Hospitalet, Spain
| | - Laura Herrero
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Marijana Todorčević
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain
| |
Collapse
|
22
|
Stenberg K, Novotny GW, Lutz TA, Mandrup-Poulsen T, Bjørnvad CR. Obesity-induced changes in gene expression in feline adipose and skeletal muscle tissue. J Anim Physiol Anim Nutr (Berl) 2023; 107:1262-1278. [PMID: 36591865 DOI: 10.1111/jpn.13802] [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: 03/21/2022] [Revised: 11/14/2022] [Accepted: 12/05/2022] [Indexed: 01/03/2023]
Abstract
Indoor-confined cats are prone to developing obesity due to a sedentary life and an energy intake exceeding energy requirements. As in humans, feline obesity decreases insulin sensitivity and increases the risk of developing feline diabetes mellitus, but the pathophysiological mechanisms are currently poorly understood. Human obesity-related metabolic alterations seem to relate to changes in the expression of genes involved in glucose metabolism, insulin action and inflammation. The objective of the current study was to investigate changes in the expression of genes relating to obesity, glucose metabolism and inflammation in cats with non-experimentally induced obesity. Biopsies from the sartorius muscle and subcutaneous adipose tissue were obtained from 73 healthy, neutered, indoor-confined domestic shorthaired cats ranging from lean to obese. Quantification of obesity-related gene expression levels relative to glyceraldehyde-3-phosphate dehydrogenase was performed by quantitative real-time polymerase chain reaction. A negative association between obesity and adiponectin expression was observed in the adipose tissue (mean ± SD; normal weight, 27.30 × 10-3 ± 77.14 × 10-3 ; overweight, 2.89 × 10-3 ± 0.38 × 10-3 and obese, 2.93 × 10-3 ± 4.20 × 10-3 , p < 0.05). In muscle, the expression of peroxisome proliferative activated receptor-γ2 and plasminogen activator inhibitor-1 was increased in the obese compared to the normal-weight cats, and resistin was increased in the normal-weight compared to the overweight cats. There were no detectable obesity-related changes in the messenger RNA levels of inflammatory cytokines. In conclusion, a possible obesity-related low-grade inflammation caused by increased expression of key proinflammatory regulators was not observed. This could imply that the development of feline obesity and ensuing insulin resistance may not be based on tissue-derived inflammation, but caused by several determining factors, many of which still need further investigation.
Collapse
Affiliation(s)
- Kathrine Stenberg
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Guy W Novotny
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Departments of Hematology/Pathology, Herlev Hospital, Herlev, Denmark
| | - Thomas A Lutz
- Institute of Veterinary Physiology, Vetsuisse-Faculty University of Zurich, Zurich, Switzerland
| | | | - Charlotte Reinhard Bjørnvad
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
23
|
Hiramoto K, Imai M, Tanaka S, Ooi K. Dementia Is Induced via the AGEs/Iba1/iNOS Pathway in Aged KK-Ay/Tajcl Mice. Life (Basel) 2023; 13:1540. [PMID: 37511915 PMCID: PMC10381697 DOI: 10.3390/life13071540] [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: 06/14/2023] [Revised: 07/07/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
The onset and exacerbation of dementia have been observed in elderly patients with type 2 diabetes. However, the underlying mechanism remains unclear. In this study, we investigated the effects of aging on the cognitive function in a mouse model of type 2 diabetes. Pathogen-free KK-Ay/TaJcl mice were used in this study. The cognitive abilities and memory declined in the mice and worsened in the 50-week-olds. The levels of advanced glycation end products (AGEs), receptor for AGE (RAGE), and Iba1 in the hippocampus were increased in the mice compared to those in the control mice. Hippocampal levels of CC-chemokine receptor 7 and inducible nitric oxide synthase, which are from M1-type macrophages that shift from microglia, were higher in KK-Ay/TaJcl mice than in control mice. Tumor necrosis factor (TNF)-α and nitric oxide (NO) levels secreted by M1-type macrophages were similarly elevated in the mice and were even higher at the age of 50 weeks. NO levels were markedly elevated in the 50-week-old mice. In contrast, differentiation of CD163 and arginase-1 did not change in both mouse types. Memory and learning declined with age in diabetic mice, and the AGEs/RAGE/M1-type macrophage/NO and TNF-α pathways played an important role in exacerbating memory and learning in those mice.
Collapse
Affiliation(s)
- Keiichi Hiramoto
- Department of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka 513-8670, Japan
| | - Masashi Imai
- Department of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka 513-8670, Japan
| | - Shota Tanaka
- Department of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka 513-8670, Japan
| | - Kazuya Ooi
- Department of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka 513-8670, Japan
| |
Collapse
|
24
|
Asakawa M, Takagi N, Hamada D, Yamasaki Y, Katsuta H. Efficacy of 3 months of additional pioglitazone treatment in type 2 diabetes patients with alcoholic fatty liver disease. Diabetol Int 2023; 14:243-251. [PMID: 37397908 PMCID: PMC10307745 DOI: 10.1007/s13340-023-00619-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/31/2023] [Indexed: 02/21/2023]
Abstract
Pioglitazone ameliorates liver dysfunction in type 2 diabetes (T2D) patients with non-alcoholic fatty liver disease (NAFLD); however, its efficacy in T2D patients with alcoholic fatty liver disease (AFLD) is unclear. Here, we conducted a retrospective single-center trial investigating whether pioglitazone ameliorates liver dysfunction in T2D patients with AFLD. T2D patients (n = 100) receiving 3 months of additional pioglitazone were divided into those with or without fatty liver (FL), and those with FL were further classified into AFLD (n = 21) and NAFLD (n = 57) groups. The effects of pioglitazone were compared across groups using medical record data on body weight changes; HbA1c, aspartate aminotransferase (AST), alanine aminotransferase (ALT), and gamma-glutamyl transpeptidase (γ-GTP) levels; and fibrosis-4 (FIB-4) index. The pioglitazone dose (mean dose: 10.6 ± 4.6 mg/day) did not affect weight gain but significantly decreased the HbA1c level in patients with or without FL (P < 0.01 and P < 0.05, respectively). The decrease in HbA1c level was significantly more pronounced in patients with FL than in those without FL (P < 0.05). In patients with FL, the HbA1c, AST, ALT, and γ-GTP levels significantly decreased after pioglitazone treatment than before (P < 0.01). The AST and ALT levels, but not the γ-GTP level, and the FIB-4 index significantly decreased after pioglitazone addition in the AFLD group, similar to that in the NAFLD group (P < 0.05 and P < 0.01, respectively). Similar effects were observed following low-dose pioglitazone treatment (≤ 7.5 mg/day) (P < 0.05) in T2D patients with AFLD and NAFLD. These results suggest that pioglitazone may be also an effective treatment option for T2D patients with AFLD.
Collapse
Affiliation(s)
- Masahiro Asakawa
- Department of Endocrinology and Metabolism, Tokyo Teishin Hospital, 2-14-23, Fujimi, Chiyoda-ku, Tokyo, Japan
| | - Noriko Takagi
- Department of Endocrinology and Metabolism, Tokyo Teishin Hospital, 2-14-23, Fujimi, Chiyoda-ku, Tokyo, Japan
| | - Daisuke Hamada
- Department of Endocrinology and Metabolism, Tokyo Teishin Hospital, 2-14-23, Fujimi, Chiyoda-ku, Tokyo, Japan
| | - Yuko Yamasaki
- Department of Endocrinology and Metabolism, Tokyo Teishin Hospital, 2-14-23, Fujimi, Chiyoda-ku, Tokyo, Japan
| | - Hidenori Katsuta
- Department of Endocrinology and Metabolism, Tokyo Teishin Hospital, 2-14-23, Fujimi, Chiyoda-ku, Tokyo, Japan
| |
Collapse
|
25
|
Jang EJ, Lee DH, Im SS, Yee J, Gwak HS. Correlation between PPARG Pro12Ala Polymorphism and Therapeutic Responses to Thiazolidinediones in Patients with Type 2 Diabetes: A Meta-Analysis. Pharmaceutics 2023; 15:1778. [PMID: 37376225 PMCID: PMC10303709 DOI: 10.3390/pharmaceutics15061778] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/09/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Background: Thiazolidinediones (TZDs) are a type of oral drug that are utilized for the treatment of type 2 diabetes mellitus (T2DM). They function by acting as agonists for a nuclear transcription factor known as peroxisome proliferator-activated receptor-gamma (PPAR-γ). TZDs, such as pioglitazone and rosiglitazone, help enhance the regulation of metabolism in individuals with T2DM by improving their sensitivity to insulin. Previous studies have suggested a relationship between the therapeutic efficacy of TZDs and the PPARG Pro12Ala polymorphism (C > G, rs1801282). However, the small sample sizes of these studies may limit their applicability in clinical settings. To address this limitation, we conducted a meta-analysis assessing the influence of the PPARG Pro12Ala polymorphism on the responsiveness of TZDs. Method: We registered our study protocol with PROSPERO, number CRD42022354577. We conducted a comprehensive search of the PubMed, Web of Science, and Embase databases, including studies published up to August 2022. We examined studies investigating the association between the PPARG Pro12Ala polymorphism and metabolic parameters such as hemoglobin A1C (HbA1C), fasting plasma glucose (FPG), triglyceride (TG), low-density lipoprotein cholesterol (LDL), high-density lipoprotein cholesterol (HDL), and total cholesterol (TC). The mean difference (MD) and 95% confidence intervals (CIs) between pre- and post-drug administration were evaluated. The quality of the studies included in the meta-analysis was assessed by using the Newcastle-Ottawa Scale (NOS) tool for cohort studies. Heterogeneity across studies was assessed by using the I2 value. An I2 value greater than 50% indicated substantial heterogeneity, and a random-effects model was used for meta-analysis. If the I2 value was below 50%, a fixed-effects model was employed instead. Both Begg's rank correlation test and Egger's regression test were performed to detect publication bias, using R Studio software. Results: Our meta-analysis incorporated 6 studies with 777 patients for blood glucose levels and 5 studies with 747 patients for lipid levels. The included studies were published between 2003 and 2016, with the majority involving Asian populations. Five of the six studies utilized pioglitazone, while the remaining study employed rosiglitazone. The quality scores, as assessed with the NOS, ranged from 8 to 9. Patients carrying the G allele exhibited a significantly greater reduction in HbA1C (MD = -0.3; 95% CI = -0.55 to -0.05; p = 0.02) and FPG (MD = -10.91; 95% CI = -19.82 to -2.01; p = 0.02) levels compared to those with the CC genotype. Furthermore, individuals with the G allele experienced a significantly larger decrease in TG levels than those with the CC genotype (MD = -26.88; 95% CI = -41.30 to -12.46; p = 0.0003). No statistically significant differences were observed in LDL (MD = 6.69; 95% CI = -0.90 to 14.29; p = 0.08), HDL (MD = 0.31; 95% CI = -1.62 to 2.23; p = 0.75), and TC (MD = 6.4; 95% CI = -0.05 to 12.84; p = 0.05) levels. No evidence of publication bias was detected based on Begg's test and Egger's test results. Conclusions: This meta-analysis reveals that patients with the Ala12 variant in the PPARG Pro12Ala polymorphism are more likely to exhibit positive responses to TZD treatment in terms of HbA1C, FPG, and TG levels compared to those with the Pro12/Pro12 genotype. These findings suggest that genotyping the PPARG Pro12Ala in diabetic patients may be advantageous for devising personalized treatment strategies, particularly for identifying individuals who are likely to respond favorably to TZDs.
Collapse
Affiliation(s)
- Eun Jeong Jang
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea; (E.J.J.); (D.H.L.); (J.Y.)
| | - Da Hoon Lee
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea; (E.J.J.); (D.H.L.); (J.Y.)
| | - Sae-Seul Im
- Graduate School of Clinical Biohealth, Ewha Womans University, Seoul 03760, Republic of Korea;
| | - Jeong Yee
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea; (E.J.J.); (D.H.L.); (J.Y.)
| | - Hye Sun Gwak
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea; (E.J.J.); (D.H.L.); (J.Y.)
| |
Collapse
|
26
|
Dana N, Ferns GA, Nedaeinia R, Haghjooy Javanmard S. Leptin signaling in breast cancer and its crosstalk with peroxisome proliferator-activated receptors α and γ. Clin Transl Oncol 2023; 25:601-610. [PMID: 36348225 DOI: 10.1007/s12094-022-02988-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 10/18/2022] [Indexed: 11/09/2022]
Abstract
Obesity may create a mitogenic microenvironment that influences tumor initiation and progression. The obesity-associated adipokine, leptin regulates energy metabolism and has been implicated in cancer development. It has been shown that some cell types other than adipocytes can express leptin and leptin receptors in tumor microenvironments. It has been shown that peroxisome proliferator-activated receptors (PPAR) agonists can affect leptin levels and vice versa leptin can affect PPARs. Activation of PPARs affects the expression of several genes involved in aspects of lipid metabolism. In addition, PPARs regulate cancer cell progression through their action on the tumor cell proliferation, metabolism, and cellular environment. Some studies have shown an association between obesity and several types of cancer, including breast cancer. There is some evidence that suggests that there is crosstalk between PPARs and leptin during the development of breast cancer. Through a systematic review of previous studies, we have reviewed the published relevant articles regarding leptin signaling in breast cancer and its crosstalk with peroxisome proliferator-activated receptors α and γ.
Collapse
Affiliation(s)
- Nasim Dana
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton and Sussex Medical School, Falmer, Brighton, BN1 9PH, Sussex, UK
| | - Reza Nedaeinia
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shaghayegh Haghjooy Javanmard
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
27
|
Calderón-DuPont D, Romero-Córdoba S, Tello JK, Espinosa A, Guerrero B, Contreras AV, Morán-Ramos S, Díaz-Villaseñor A. Impaired white adipose tissue fatty acid metabolism in mice fed a high-fat diet worsened by arsenic exposure, primarily affecting retroperitoneal adipose tissue. Toxicol Appl Pharmacol 2023; 468:116428. [PMID: 36801214 DOI: 10.1016/j.taap.2023.116428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/10/2023] [Accepted: 02/11/2023] [Indexed: 02/17/2023]
Abstract
Fatty acid (FA) metabolism dysfunction of white adipose tissue (WAT) underlies obesity and insulin resistance in response to high calorie intake and/or endocrine-disrupting chemicals (EDCs), among other factors. Arsenic is an EDC that has been associated with metabolic syndrome and diabetes. However, the combined effect of a high-fat diet (HFD) and arsenic exposure on WAT FA metabolism has been little studied. FA metabolism was evaluated in visceral (epididymal and retroperitoneal) and subcutaneous WAT of C57BL/6 male mice fed control or HFD (12 and 40% kcal fat, respectively) for 16 weeks together with an environmentally relevant chronic arsenic exposure through drinking water (100 μg/l) during the second half of the study. In mice fed HFD, arsenic potentiated the increase of serum markers of selective insulin resistance in WAT and fatty acid re-esterification and the decrease in the lipolysis index. Retroperitoneal was the WAT most affected, where the combination of arsenic and HFD in contrast to HFD, generated higher weight, larger adipocytes, increased triglyceride content, and decreased fasting stimulated lipolysis evidenced by lower phosphorylation of HSL and perilipin. At the transcriptional level, arsenic in mice fed either diet downregulated genes involved in fatty acid uptake (LPL, CD36), oxidation (PPARα, CPT1), lipolysis (ADRß3) and glycerol transport (AQP7 and AQP9). Additionally, arsenic potentiated hyperinsulinemia induced by HFD, despite a slight increase in weight gain and food efficiency. Thus, the second hit of arsenic in sensitized mice by HFD worsens fatty acid metabolism impairment in WAT, mainly retroperitoneal, along with an exacerbated insulin resistance phenotype.
Collapse
Affiliation(s)
- Diana Calderón-DuPont
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City 045010, Mexico; Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City 045010, Mexico
| | - Sandra Romero-Córdoba
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City 045010, Mexico; Departamento de Bioquímica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14000, Mexico
| | - Jessica K Tello
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City 045010, Mexico; Maestría en Nutrición Clínica, Universidad Anáhuac Campus Norte, Estado de México 52786, Mexico
| | - Aranza Espinosa
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City 045010, Mexico; Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City 045010, Mexico
| | - Brenda Guerrero
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City 045010, Mexico; Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City 045010, Mexico
| | - Alejandra V Contreras
- Laboratorio de Nutrigenética y Nutrigenómica, Instituto Nacional de Medicina Genόmica (INMEGEN), Mexico City 14609, Mexico; Translational Molecular Biomarkers, Merck & Co., Inc, Rahway, NJ, USA
| | - Sofia Morán-Ramos
- Unidad de Genόmica de Poblaciones Aplicada a la Salud, Facultad de Química, UNAM/Instituto Nacional de Medicina Genόmica (INMEGEN), Mexico City 14609, Mexico; Departamento de Alimentos y Biotecnología, Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City 045010, Mexico
| | - Andrea Díaz-Villaseñor
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City 045010, Mexico.
| |
Collapse
|
28
|
Iizaka T, Kodama E, Mikura K, Iida T, Imai H, Hashizume M, Kigawa Y, Sugisawa C, Tadokoro R, Endo K, Otsuka F, Isoda M, Ebihara K, Ishibashi S, Nagasaka S. Clinical characteristics and efficacy of pioglitazone in a Japanese patient with familial partial lipodystrophy due to peroxisome proliferator-activated receptor γ gene mutation. Endocr J 2023; 70:69-76. [PMID: 36171144 DOI: 10.1507/endocrj.ej22-0140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Familial partial lipodystrophy (FPLD) 3 is a rare genetic disorder caused by peroxisome proliferator-activated receptor γ gene (PPARG) mutations. Most cases have been reported in Western patients. Here, we describe a first pedigree of FPLD 3 in Japanese. The proband was a 51-year-old woman. She was diagnosed with fatty liver at age 32 years, dyslipidemia at age 37 years, and diabetes mellitus at age 41 years. Her body mass index was 18.5 kg/m2, and body fat percentage was 19.2%. On physical examination, she had less subcutaneous fat in the upper limbs than in other sites. On magnetic resonance imaging, atrophy of subcutaneous adipose tissue was seen in the upper limbs and lower legs. Fasting serum C-peptide immunoreactivity was high (3.4 ng/mL), and the plasma glucose disappearance rate was low (2.07%/min) on an insulin tolerance test, both suggesting apparent insulin resistance. The serum total adiponectin level was low (2.3 μg/mL). Mild fatty liver was seen on abdominal computed tomography. On genetic analysis, a P495L mutation in PPARG was identified. The same mutation was also seen in her father, who had non-obese diabetes mellitus, and FPLD 3 was diagnosed. Modest increases in body fat and serum total adiponectin were seen with pioglitazone treatment. Attention should be paid to avoid overlooking lipodystrophy syndromes even in non-obese diabetic patients if they show features of insulin resistance.
Collapse
Affiliation(s)
- Toru Iizaka
- Division of Diabetes, Metabolism and Endocrinology, Showa University Fujigaoka Hospital, Kanagawa 227-8501, Japan
| | - Eriko Kodama
- Division of Diabetes, Metabolism and Endocrinology, Showa University Fujigaoka Hospital, Kanagawa 227-8501, Japan
| | - Kentaro Mikura
- Division of Diabetes, Metabolism and Endocrinology, Showa University Fujigaoka Hospital, Kanagawa 227-8501, Japan
| | - Tatsuya Iida
- Division of Diabetes, Metabolism and Endocrinology, Showa University Fujigaoka Hospital, Kanagawa 227-8501, Japan
| | - Hideyuki Imai
- Division of Diabetes, Metabolism and Endocrinology, Showa University Fujigaoka Hospital, Kanagawa 227-8501, Japan
| | - Mai Hashizume
- Division of Diabetes, Metabolism and Endocrinology, Showa University Fujigaoka Hospital, Kanagawa 227-8501, Japan
| | - Yasuyoshi Kigawa
- Division of Diabetes, Metabolism and Endocrinology, Showa University Fujigaoka Hospital, Kanagawa 227-8501, Japan
| | - Chiho Sugisawa
- Division of Diabetes, Metabolism and Endocrinology, Showa University Fujigaoka Hospital, Kanagawa 227-8501, Japan
| | - Rie Tadokoro
- Division of Diabetes, Metabolism and Endocrinology, Showa University Fujigaoka Hospital, Kanagawa 227-8501, Japan
| | - Kei Endo
- Division of Diabetes, Metabolism and Endocrinology, Showa University Fujigaoka Hospital, Kanagawa 227-8501, Japan
| | - Fumiko Otsuka
- Division of Diabetes, Metabolism and Endocrinology, Showa University Fujigaoka Hospital, Kanagawa 227-8501, Japan
| | - Masayo Isoda
- Division of Endocrinology and Metabolism, Jichi Medical University, Tochigi 329-0498, Japan
| | - Ken Ebihara
- Division of Endocrinology and Metabolism, Jichi Medical University, Tochigi 329-0498, Japan
| | - Shun Ishibashi
- Division of Endocrinology and Metabolism, Jichi Medical University, Tochigi 329-0498, Japan
| | - Shoichiro Nagasaka
- Division of Diabetes, Metabolism and Endocrinology, Showa University Fujigaoka Hospital, Kanagawa 227-8501, Japan
| |
Collapse
|
29
|
Takahashi J, Takahashi N, Tadaishi M, Shimizu M, Kobayashi-Hattori K. Valerenic Acid Promotes Adipocyte Differentiation, Adiponectin Production, and Glucose Uptake via Its PPARγ Ligand Activity. ACS OMEGA 2022; 7:48113-48120. [PMID: 36591200 PMCID: PMC9798764 DOI: 10.1021/acsomega.2c06120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 11/28/2022] [Indexed: 06/17/2023]
Abstract
Although valerenic acid (VA) is an important marker compound for quantitative assessment of Valeriana officinalis products, little is known about its potential effects on adipocytes. We investigated the effects of VA on adipocyte differentiation, adiponectin production, and glucose uptake using 3T3-L1 adipocytes. The results showed that VA promoted adipocyte differentiation and increased the gene expression of adipogenesis and glucose uptake-related proteins, including peroxisome proliferator-activated receptor gamma (PPARγ), cytosine-cytosine-adenosine-adenosine-thymidine enhancer binding protein alpha (C/EBPα), adiponectin, and glucose transporter 4 (GLUT4). Additionally, cell cultures treated with VA had elevated adiponectin secretion and glucose uptake. The PPARγ luciferase assay indicated VA as a partial agonist of PPARγ, while the analysis using its antagonist, GW9662, and a docking simulation between PPARγ and VA revealed the binding site of VA as likely adjacent to the Ω loop pocket of PPARγ. Taken together, these results demonstrate that VA acts as a PPARγ partial agonist to promote adipocyte differentiation, adiponectin production, and glucose uptake.
Collapse
Affiliation(s)
- Jun Takahashi
- Department
of Nutritional Science, Faculty of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo 156-8502, Japan
| | - Nobuyuki Takahashi
- Department
of Nutritional Science and Food Safety, Faculty of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo 156-8502, Japan
| | - Miki Tadaishi
- Department
of Nutritional Science, Faculty of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo 156-8502, Japan
| | - Makoto Shimizu
- Department
of Nutritional Science, Faculty of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo 156-8502, Japan
| | - Kazuo Kobayashi-Hattori
- Department
of Nutritional Science, Faculty of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo 156-8502, Japan
| |
Collapse
|
30
|
Mukherjee AG, Wanjari UR, Gopalakrishnan AV, Katturajan R, Kannampuzha S, Murali R, Namachivayam A, Ganesan R, Renu K, Dey A, Vellingiri B, Prince SE. Exploring the Regulatory Role of ncRNA in NAFLD: A Particular Focus on PPARs. Cells 2022; 11:3959. [PMID: 36552725 PMCID: PMC9777112 DOI: 10.3390/cells11243959] [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: 09/27/2022] [Revised: 11/29/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022] Open
Abstract
Liver diseases are responsible for global mortality and morbidity and are a significant cause of death worldwide. Consequently, the advancement of new liver disease targets is of great interest. Non-coding RNA (ncRNA), such as microRNA (miRNA) and long ncRNA (lncRNA), has been proven to play a significant role in the pathogenesis of virtually all acute and chronic liver disorders. Recent studies demonstrated the medical applications of miRNA in various phases of hepatic pathology. PPARs play a major role in regulating many signaling pathways involved in various metabolic disorders. Non-alcoholic fatty liver disease (NAFLD) is the most prevalent form of chronic liver disease in the world, encompassing a spectrum spanning from mild steatosis to severe non-alcoholic steatohepatitis (NASH). PPARs were found to be one of the major regulators in the progression of NAFLD. There is no recognized treatment for NAFLD, even though numerous clinical trials are now underway. NAFLD is a major risk factor for developing hepatocellular carcinoma (HCC), and its frequency increases as obesity and diabetes become more prevalent. Reprogramming anti-diabetic and anti-obesity drugs is an effective therapy option for NAFLD and NASH. Several studies have also focused on the role of ncRNAs in the pathophysiology of NAFLD. The regulatory effects of these ncRNAs make them a primary target for treatments and as early biomarkers. In this study, the main focus will be to understand the regulation of PPARs through ncRNAs and their role in NAFLD.
Collapse
Affiliation(s)
- Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Ramkumar Katturajan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Sandra Kannampuzha
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Reshma Murali
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Arunraj Namachivayam
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Raja Ganesan
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 24252, Republic of Korea
| | - Kaviyarasi Renu
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata 700073, West Bengal, India
| | - Balachandar Vellingiri
- Stem Cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda 151401, Punjab, India
| | - Sabina Evan Prince
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| |
Collapse
|
31
|
Vasandani C, Li X, Sekizkardes H, Brown RJ, Garg A. Phenotypic Differences Among Familial Partial Lipodystrophy Due to LMNA or PPARG Variants. J Endocr Soc 2022; 6:bvac155. [PMID: 36397776 PMCID: PMC9664976 DOI: 10.1210/jendso/bvac155] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Indexed: 11/19/2022] Open
Abstract
Context Despite several reports of familial partial lipodystrophy (FPLD) type 2 (FPLD2) due to heterozygous LMNA variants and FPLD3 due to PPARG variants, the phenotypic differences among them remain unclear. Objective To compare the body fat distribution, metabolic parameters, and prevalence of metabolic complications between FPLD3 and FPLD2. Methods A retrospective, cross-sectional comparison of patients from 2 tertiary referral centers-UT Southwestern Medical Center and the National Institute of Diabetes and Digestive and Kidney Diseases. A total of 196 females and 59 males with FPLD2 (age 2-86 years) and 28 females and 4 males with FPLD3 (age 9-72 years) were included. The main outcome measures were skinfold thickness, regional body fat by dual-energy X-ray absorptiometry (DXA), metabolic variables, and prevalence of diabetes mellitus and hypertriglyceridemia. Results Compared with subjects with FPLD2, subjects with FPLD3 had significantly increased prevalence of hypertriglyceridemia (66% vs 84%) and diabetes (44% vs 72%); and had higher median fasting serum triglycerides (208 vs 255 mg/dL), and mean hemoglobin A1c (6.4% vs 7.5%). Compared with subjects with FPLD2, subjects with FPLD3 also had significantly higher mean upper limb fat (21% vs 27%) and lower limb fat (16% vs 21%) on DXA and increased median skinfold thickness at the anterior thigh (5.8 vs 11.3 mm), calf (4 vs 6 mm), triceps (5.5 vs 7.5 mm), and biceps (4.3 vs 6.8 mm). Conclusion Compared with subjects with FPLD2, subjects with FPLD3 have milder lipodystrophy but develop more severe metabolic complications, suggesting that the remaining adipose tissue in subjects with FPLD3 may be dysfunctional or those with mild metabolic disease are underrecognized.
Collapse
Affiliation(s)
- Chandna Vasandani
- Division of Nutrition and Metabolic Diseases and the Center for Human Nutrition, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xilong Li
- Department of Population and Data Sciences, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Hilal Sekizkardes
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rebecca J Brown
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Abhimanyu Garg
- Division of Nutrition and Metabolic Diseases and the Center for Human Nutrition, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
32
|
Przybycień P, Gąsior-Perczak D, Placha W. Cannabinoids and PPAR Ligands: The Future in Treatment of Polycystic Ovary Syndrome Women with Obesity and Reduced Fertility. Cells 2022; 11:cells11162569. [PMID: 36010645 PMCID: PMC9406585 DOI: 10.3390/cells11162569] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 08/13/2022] [Accepted: 08/17/2022] [Indexed: 11/21/2022] Open
Abstract
Cannabinoids (CBs) are used to treat chronic pain, chemotherapy-induced nausea and vomiting, and multiple sclerosis spasticity. Recently, the medicinal use of CBs has attracted increasing interest as a new therapeutic in many diseases. Data indicate a correlation between CBs and PPARs via diverse mechanisms. Both the endocannabinoid system (ECS) and peroxisome proliferator-activated receptors (PPARs) may play a significant role in PCOS and PCOS related disorders, especially in disturbances of glucose-lipid metabolism as well as in obesity and fertility. Taking into consideration the ubiquity of PCOS in the human population, it seems indispensable to search for new potential therapeutic targets for this condition. The aim of this review is to examine the relationship between metabolic disturbances and obesity in PCOS pathology. We discuss current and future therapeutic interventions for PCOS and related disorders, with emphasis on the metabolic pathways related to PCOS pathophysiology. The link between the ECS and PPARs is a promising new target for PCOS, and we examine this relationship in depth.
Collapse
Affiliation(s)
- Piotr Przybycień
- Chair of Medical Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, 31-034 Krakow, Poland
- Endocrinology Clinic, Holycross Cancer Centre, 25-734 Kielce, Poland
| | - Danuta Gąsior-Perczak
- Endocrinology Clinic, Holycross Cancer Centre, 25-734 Kielce, Poland
- Collegium Medicum, Jan Kochanowski University, 25-317 Kielce, Poland
| | - Wojciech Placha
- Chair of Medical Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, 31-034 Krakow, Poland
- Correspondence: ; Tel.: +48-12-422-74-00
| |
Collapse
|
33
|
Zhang ZB, Sinha J, Bahrami-Nejad Z, Teruel MN. The circadian clock mediates daily bursts of cell differentiation by periodically restricting cell-differentiation commitment. Proc Natl Acad Sci U S A 2022; 119:e2204470119. [PMID: 35939672 PMCID: PMC9388110 DOI: 10.1073/pnas.2204470119] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 06/20/2022] [Indexed: 11/18/2022] Open
Abstract
Most mammalian cells have an intrinsic circadian clock that coordinates metabolic activity with the daily rest and wake cycle. The circadian clock is known to regulate cell differentiation, but how continuous daily oscillations of the internal clock can control a much longer, multiday differentiation process is not known. Here, we simultaneously monitor circadian clock and adipocyte-differentiation progression live in single cells. Strikingly, we find a bursting behavior in the cell population whereby individual preadipocytes commit to differentiate primarily during a 12-h window each day, corresponding to the time of rest. Daily gating occurs because cells irreversibly commit to differentiate within only a few hours, which is much faster than the rest phase and the overall multiday differentiation process. The daily bursts in differentiation commitment result from a differentiation-stimulus driven variable and slow increase in expression of PPARG, the master regulator of adipogenesis, overlaid with circadian boosts in PPARG expression driven by fast, clock-driven PPARG regulators such as CEBPA. Our finding of daily bursts in cell differentiation only during the circadian cycle phase corresponding to evening in humans is broadly relevant, given that most differentiating somatic cells are regulated by the circadian clock. Having a restricted time each day when differentiation occurs may open therapeutic strategies to use timed treatment relative to the clock to promote tissue regeneration.
Collapse
Affiliation(s)
- Zhi-Bo Zhang
- Department of Biochemistry, Weill Cornell Medical College of Cornell University, New York, NY 10065
- The Ira & Gale Drukier Institute of Children’s Health, Weill Cornell Medical College of Cornell University, New York, NY 10065
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305
| | - Joydeb Sinha
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305
| | - Zahra Bahrami-Nejad
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305
| | - Mary N. Teruel
- Department of Biochemistry, Weill Cornell Medical College of Cornell University, New York, NY 10065
- The Ira & Gale Drukier Institute of Children’s Health, Weill Cornell Medical College of Cornell University, New York, NY 10065
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305
- Weill Center for Metabolic Health, Division of Endocrinology, Diabetes, and Metabolism, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College of Cornell University, New York, NY 10065
- Department of Bioengineering, Stanford University, Stanford, CA 94305
| |
Collapse
|
34
|
Effects of a Phosphodiesterase inhibitor on the Browning of Adipose Tissue in Mice. Biomedicines 2022; 10:biomedicines10081852. [PMID: 36009398 PMCID: PMC9405663 DOI: 10.3390/biomedicines10081852] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/19/2022] [Accepted: 07/25/2022] [Indexed: 11/23/2022] Open
Abstract
Cilostazol is a selective inhibitor of phosphodiesterase type 3 (PDE3) that increases intracellular cyclic adenosine monophosphate (cAMP), which plays a critical role in the development of the beige phenotype and the activation of its thermogenic program in white adipose tissue (WAT). We investigated the metabolic effects of PDE3B inhibition with cilostazol treatment in the adipose tissue of high-fat diet (HFD)-fed mice. Seven-week-old male C57BL/6J mice were randomly assigned to either the cilostazol or control group. The control group was divided into two groups: the chow diet and HFD. The expression of uncoupling Protein 1 (UCP1) and other brown adipocyte markers was compared. In the HFD-fed cilostazol group, C57BL/6J mice displayed improvements in systemic metabolism, including improved glucose tolerance and lipid profile, but only modest effects on body weight were observed. In the visceral WAT of HFD-fed cilostazol-treated mice, cAMP/protein kinase A (PKA) signaling pathways were activated, resulting in the “browning” phenotype, smaller fat deposits, and enhanced mRNA expression of UCP1 and other brown adipocyte markers. PDE3B appears to be an important regulator of lipid metabolism, insulin sensitivity, and thermogenic programs in adipose tissues. An increase in intracellular cAMP via PDE3B inhibition with cilostazol treatment promoted the browning of visceral WAT.
Collapse
|
35
|
Tang X, Zhou H, Yan H, Niimi M, Fan J. Is apoCIII-Lowering A Double-Edged Sword? J Atheroscler Thromb 2022; 29:1117-1124. [PMID: 35545534 PMCID: PMC9252641 DOI: 10.5551/jat.le002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Affiliation(s)
- Xiangming Tang
- Department of Molecular Pathology, Faculty of Medicine, Interdisciplinary Graduate School of Medicine, University of Yamanashi
| | - Huanjin Zhou
- School of Biotechnology and Health Sciences, Wuyi University
| | - Haizhao Yan
- Department of Molecular Pathology, Faculty of Medicine, Interdisciplinary Graduate School of Medicine, University of Yamanashi.,Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences
| | - Manabu Niimi
- Department of Molecular Pathology, Faculty of Medicine, Interdisciplinary Graduate School of Medicine, University of Yamanashi
| | - Jianglin Fan
- Department of Molecular Pathology, Faculty of Medicine, Interdisciplinary Graduate School of Medicine, University of Yamanashi.,School of Biotechnology and Health Sciences, Wuyi University
| |
Collapse
|
36
|
Müller C, Zidek LM, Eichwald S, Kortman G, Koster MH, Calkhoven CF. Enhanced C/EBPβ function promotes hypertrophic versus hyperplastic fat tissue growth and prevents steatosis in response to high-fat diet feeding. eLife 2022; 11:e62625. [PMID: 35451956 PMCID: PMC9071262 DOI: 10.7554/elife.62625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 04/08/2022] [Indexed: 11/30/2022] Open
Abstract
Chronic obesity is correlated with severe metabolic and cardiovascular diseases as well as with an increased risk for developing cancers. Obesity is usually characterized by fat accumulation in enlarged - hypertrophic - adipocytes that are a source of inflammatory mediators, which promote the development and progression of metabolic disorders. Yet, in certain healthy obese individuals, fat is stored in metabolically more favorable hyperplastic fat tissue that contains an increased number of smaller adipocytes that are less inflamed. In a previous study, we demonstrated that loss of the inhibitory protein-isoform C/EBPβ-LIP and the resulting augmented function of the transactivating isoform C/EBPβ-LAP promotes fat metabolism under normal feeding conditions and expands health- and lifespan in mice. Here, we show that in mice on a high-fat diet, LIP-deficiency results in adipocyte hyperplasia associated with reduced inflammation and metabolic improvements. Furthermore, fat storage in subcutaneous depots is significantly enhanced specifically in LIP-deficient male mice. Our data identify C/EBPβ as a regulator of adipocyte fate in response to increased fat intake, which has major implications for metabolic health and aging.
Collapse
Affiliation(s)
- Christine Müller
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of GroningenGroningenNetherlands
| | - Laura M Zidek
- Leibniz Institute on Aging - Fritz Lipmann InstituteJenaGermany
| | | | - Gertrud Kortman
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of GroningenGroningenNetherlands
| | - Mirjam H Koster
- Division Molecular Genetics, Department of Pediatrics, University Medical Center Groningen, University of GroningenGroningenNetherlands
| | - Cornelis F Calkhoven
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of GroningenGroningenNetherlands
- Leibniz Institute on Aging - Fritz Lipmann InstituteJenaGermany
| |
Collapse
|
37
|
Fryklund C, Morén B, Neuhaus M, Periwal V, Stenkula KG. Rosiglitazone treatment enhances intracellular actin dynamics and glucose transport in hypertrophic adipocytes. Life Sci 2022; 299:120537. [PMID: 35398016 DOI: 10.1016/j.lfs.2022.120537] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 03/28/2022] [Accepted: 04/02/2022] [Indexed: 10/18/2022]
Abstract
AIMS To accommodate surplus energy, adipose tissue expands by increasing both adipose cell size (hypertrophy) and cell number (hyperplasia). Enlarged, hypertrophic adipocytes are known to have reduced insulin response and impaired glucose transport, which negatively influence whole-body glucose homeostasis. Rosiglitazone is a peroxisome proliferator-activated receptor gamma (PPARγ) agonist, known to stimulate hyperplasia and to efficiently improve insulin sensitivity. Still, a limited amount of research has investigated the effects of rosiglitazone in mature, hypertrophic adipocytes. Therefore, the objective of this study was to examine rosiglitazone's effect on insulin-stimulated glucose uptake in hypertrophic adipocytes. MAIN METHODS C57BL/6J male mice were subjected to 2 weeks of high-fat diet (HFD) followed by 1 week of HFD combined with daily administration of rosiglitazone (10 mg/kg). Adipose cell-size distribution and gene expression were analysed in intact adipose tissue, and glucose uptake, insulin response, and protein expression were examined using primary adipocytes isolated from epididymal and inguinal adipose tissue. KEY FINDINGS HFD-feeding induced an accumulation of hypertrophic adipocytes, which was not affected by rosiglitazone-treatment. Still, rosiglitazone efficiently improved insulin-stimulated glucose transport without restoring insulin signaling or GLUT4 expression in similar-sized adipocytes. This improvement occurred concurrently with extracellular matrix remodelling and restored intracellular levels of targets involved in actin turnover. SIGNIFICANCE These results demonstrate that rosiglitazone improves glucose transport in hypertrophic adipocytes, and highlights the importance of the cytoskeleton and extracellular matrix as potential therapeutic targets.
Collapse
Affiliation(s)
- Claes Fryklund
- Department of Experimental Medical Science, Lund University, Sweden.
| | - Björn Morén
- Department of Experimental Medical Science, Lund University, Sweden
| | | | - Vipul Periwal
- Laboratory of Biological Modeling, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, USA
| | - Karin G Stenkula
- Department of Experimental Medical Science, Lund University, Sweden
| |
Collapse
|
38
|
Bilal M, Nawaz A, Kado T, Aslam MR, Igarashi Y, Nishimura A, Watanabe Y, Kuwano T, Liu J, Miwa H, Era T, Ikuta K, Imura J, Yagi K, Nakagawa T, Fujisaka S, Tobe K. Fate of adipocyte progenitors during adipogenesis in mice fed a high-fat diet. Mol Metab 2021; 54:101328. [PMID: 34562641 PMCID: PMC8495176 DOI: 10.1016/j.molmet.2021.101328] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/09/2021] [Accepted: 08/20/2021] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE Expansion of adipose tissue during obesity through the recruitment of newly generated adipocytes (hyperplasia) is metabolically healthy, whereas that through the enlargement of pre-existing adipocytes (hypertrophy) leads to metabolic complications. Accumulating evidence from genetic fate mapping studies suggests that in animal models receiving a high-fat diet (HFD), only adipocyte progenitors (APs) in gonadal white adipose tissue (gWAT) have proliferative potential. However, the proliferative potential and differentiating capacity of APs in the inguinal WAT (iWAT) of male mice remains controversial. The objective of this study was to investigate the proliferative and adipogenic potential of APs in the iWAT of HFD-fed male mice. METHODS We generated PDGFRα-GFP-Cre-ERT2/tdTomato (KI/td) mice and traced PDGFRα-positive APs in male mice fed HFD for 8 weeks. We performed a comprehensive phenotypic analysis, including the histology, immunohistochemistry, flow cytometry, and gene expression analysis, of KI/td mice fed HFD. RESULTS Contrary to the findings of others, we found an increased number of newly generated tdTomato+ adipocytes in the iWAT of male mice, which was smaller than that observed in the gWAT. We found that in male mice, the iWAT has more proliferating tdTomato+ APs than the gWAT. We also found that tdTomato+ APs showed a higher expression of Dpp4 and Pi16 than tdTomato- APs, and the expression of these genes was significantly higher in the iWAT than in the gWAT of mice fed HFD for 8 weeks. Collectively, our results reveal that HFD feeding induces the proliferation of tdTomato+ APs in the iWAT of male mice. CONCLUSION In male mice, compared with gWAT, iWAT undergoes hyperplasia in response to 8 weeks of HFD feeding through the recruitment of newly generated adipocytes due to an abundance of APs with a high potential for proliferation and differentiation.
Collapse
Affiliation(s)
- Muhammad Bilal
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Allah Nawaz
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan; Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan.
| | - Tomonobu Kado
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Muhammad Rahil Aslam
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Yoshiko Igarashi
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Ayumi Nishimura
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Yoshiyuki Watanabe
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Takahide Kuwano
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Jianhui Liu
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Hiroyuki Miwa
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Takumi Era
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Koichi Ikuta
- Department of Virus Research, Laboratory of Immune Regulation, Institute of Frontier Life and Medical Sciences, Kyoto University, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Johji Imura
- Department of Diagnostic Pathology, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Kunimasa Yagi
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Takashi Nakagawa
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Shiho Fujisaka
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Kazuyuki Tobe
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan.
| |
Collapse
|
39
|
Miyachi H. Structural Biology-Based Exploration of Subtype-Selective Agonists for Peroxisome Proliferator-Activated Receptors. Int J Mol Sci 2021; 22:ijms22179223. [PMID: 34502131 PMCID: PMC8430769 DOI: 10.3390/ijms22179223] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 12/17/2022] Open
Abstract
Progress in understanding peroxisome proliferator-activated receptor (PPAR) subtypes as nuclear receptors that have pleiotropic effects on biological responses has enabled the exploration of new subtype-selective PPAR ligands. Such ligands are useful chemical biology/pharmacological tools to investigate the functions of PPARs and are also candidate drugs for the treatment of PPAR-mediated diseases, such as metabolic syndrome, inflammation and cancer. This review summarizes our medicinal chemistry research of more than 20 years on the design, synthesis, and pharmacological evaluation of subtype-selective PPAR agonists, which has been based on two working hypotheses, the ligand superfamily concept and the helix 12 (H12) holding induction concept. X-ray crystallographic analyses of our agonists complexed with each PPAR subtype validate our working hypotheses.
Collapse
Affiliation(s)
- Hiroyuki Miyachi
- Lead Exploration Unit, Drug Discovery Initiative, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
40
|
Allium-Derived Compound Propyl Propane Thiosulfonate (PTSO) Attenuates Metabolic Alterations in Mice Fed a High-Fat Diet through Its Anti-Inflammatory and Prebiotic Properties. Nutrients 2021; 13:nu13082595. [PMID: 34444755 PMCID: PMC8400710 DOI: 10.3390/nu13082595] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 07/22/2021] [Accepted: 07/25/2021] [Indexed: 12/30/2022] Open
Abstract
Background: Propyl propane thiosulfonate (PTSO) is an organosulfur compound from Allium spp. that has shown interesting antimicrobial properties and immunomodulatory effects in different experimental models. In this sense, our aim was to evaluate its effect on an experimental model of obesity, focusing on inflammatory and metabolic markers and the gut microbiota. Methods and results: Mice were fed a high-fat diet and orally treated with different doses of PTSO (0.1, 0.5 and 1 mg/kg/day) for 5 weeks. PTSO lessened the weight gain and improved the plasma markers associated with glucose and lipid metabolisms. PTSO also attenuated obesity-associated systemic inflammation, reducing the immune cell infiltration and, thus, the expression of pro-inflammatory cytokines in adipose and hepatic tissues (Il-1ẞ, Il-6, Tnf-α, Mcp-1, Jnk-1, Jnk-2, Leptin, Leptin R, Adiponectin, Ampk, Ppar-α, Ppar-γ, Glut-4 and Tlr-4) and improving the expression of different key elements for gut barrier integrity (Muc-2, Muc-3, Occludin, Zo-1 and Tff-3). Additionally, these effects were connected to a regulation of the gut microbiome, which was altered by the high-fat diet. Conclusion: Allium-derived PTSO can be considered a potential new tool for the treatment of metabolic syndrome.
Collapse
|
41
|
Trans-palmitoleic acid reduces adiposity via increased lipolysis in a rodent model of diet-induced obesity. Br J Nutr 2021; 127:801-809. [PMID: 33958011 DOI: 10.1017/s0007114521001501] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Obesity is defined as increased adiposity, which leads to metabolic disease. The growth of adipose tissue depends on its capacity to expand through hyperplasia or hypertrophy, in order to buffer energy surplus. Also, during the establishment of obesity, adipose tissue expansion reflects adipose lipid metabolism (lipogenesis and/or lipolysis). It is well known that dietary factors can modify lipid metabolism promoting or preventing the development of metabolic abnormalities that concur with obesity. Trans-palmitoleic acid (TP), a biomarker of dairy consumption, has been associated with reduced adiposity in clinical studies. Thus, we aimed to evaluate the effect of TP over adiposity and lipid metabolism-related genes in a rodent model of diet-induced obesity (DIO). To fulfil this aim, we fed C57BL/6 mice with a Control or a High-Fat diet, added with or without TP (3 g/kg diet), during 11 weeks. Body weight and food intake were monitored, fat pads were weighted, histology of visceral adipose tissue was analysed and lipid metabolism-related gene expression was explored by qPCR. Results show that TP consumption prevented weight gain induced by high-fat diet, reduced visceral adipose tissue weight and adipocyte size, while increasing the expression of lipolytic molecules. In conclusion, we show for the first time that TP influences adipose tissue metabolism, specifically lipolysis, resulting in decreased adiposity and reduced adipocyte size in a DIO mice model.
Collapse
|
42
|
AGMO Inhibitor Reduces 3T3-L1 Adipogenesis. Cells 2021; 10:cells10051081. [PMID: 34062826 PMCID: PMC8147360 DOI: 10.3390/cells10051081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/20/2021] [Accepted: 04/28/2021] [Indexed: 12/20/2022] Open
Abstract
Alkylglycerol monooxygenase (AGMO) is a tetrahydrobiopterin (BH4)-dependent enzyme with major expression in the liver and white adipose tissue that cleaves alkyl ether glycerolipids. The present study describes the disclosure and biological characterization of a candidate compound (Cp6), which inhibits AGMO with an IC50 of 30–100 µM and 5–20-fold preference of AGMO relative to other BH4-dependent enzymes, i.e., phenylalanine-hydroxylase and nitric oxide synthase. The viability and metabolic activity of mouse 3T3-L1 fibroblasts, HepG2 human hepatocytes and mouse RAW264.7 macrophages were not affected up to 10-fold of the IC50. However, Cp6 reversibly inhibited the differentiation of 3T3-L1 cells towards adipocytes, in which AGMO expression was upregulated upon differentiation. Cp6 reduced the accumulation of lipid droplets in adipocytes upon differentiation and in HepG2 cells exposed to free fatty acids. Cp6 also inhibited IL-4-driven differentiation of RAW264.7 macrophages towards M2-like macrophages, which serve as adipocyte progenitors in adipose tissue. Collectively, the data suggest that pharmacologic AGMO inhibition may affect lipid storage.
Collapse
|
43
|
Obesity and aging: Molecular mechanisms and therapeutic approaches. Ageing Res Rev 2021; 67:101268. [PMID: 33556548 DOI: 10.1016/j.arr.2021.101268] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 01/19/2021] [Accepted: 02/02/2021] [Indexed: 02/08/2023]
Abstract
The epidemic of obesity is a major challenge for health policymakers due to its far-reaching effects on population health and potentially overwhelming financial burden on healthcare systems. Obesity is associated with an increased risk of developing acute and chronic diseases, including hypertension, stroke, myocardial infarction, cardiovascular disease, diabetes, and cancer. Interestingly, the metabolic dysregulation associated with obesity is similar to that observed in normal aging, and substantial evidence suggests the potential of obesity to accelerate aging. Therefore, understanding the mechanism of fat tissue dysfunction in obesity could provide insights into the processes that contribute to the metabolic dysfunction associated with the aging process. Here, we review the molecular and cellular mechanisms underlying both obesity and aging, and how obesity and aging can predispose individuals to chronic health complications. The potential of lifestyle and pharmacological interventions to counter obesity and obesity-related pathologies, as well as aging, is also addressed.
Collapse
|
44
|
Dixon ED, Nardo AD, Claudel T, Trauner M. The Role of Lipid Sensing Nuclear Receptors (PPARs and LXR) and Metabolic Lipases in Obesity, Diabetes and NAFLD. Genes (Basel) 2021; 12:genes12050645. [PMID: 33926085 PMCID: PMC8145571 DOI: 10.3390/genes12050645] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/23/2021] [Accepted: 04/23/2021] [Indexed: 12/11/2022] Open
Abstract
Obesity and type 2 diabetes mellitus (T2DM) are metabolic disorders characterized by metabolic inflexibility with multiple pathological organ manifestations, including non-alcoholic fatty liver disease (NAFLD). Nuclear receptors are ligand-dependent transcription factors with a multifaceted role in controlling many metabolic activities, such as regulation of genes involved in lipid and glucose metabolism and modulation of inflammatory genes. The activity of nuclear receptors is key in maintaining metabolic flexibility. Their activity depends on the availability of endogenous ligands, like fatty acids or oxysterols, and their derivatives produced by the catabolic action of metabolic lipases, most of which are under the control of nuclear receptors. For example, adipose triglyceride lipase (ATGL) is activated by peroxisome proliferator-activated receptor γ (PPARγ) and conversely releases fatty acids as ligands for PPARα, therefore, demonstrating the interdependency of nuclear receptors and lipases. The diverse biological functions and importance of nuclear receptors in metabolic syndrome and NAFLD has led to substantial effort to target them therapeutically. This review summarizes recent findings on the roles of lipases and selected nuclear receptors, PPARs, and liver X receptor (LXR) in obesity, diabetes, and NAFLD.
Collapse
Affiliation(s)
| | | | | | - Michael Trauner
- Correspondence: ; Tel.: +43-140-4004-7410; Fax: +43-14-0400-4735
| |
Collapse
|
45
|
de Paula K, Santos JC, Mafud AC, Nascimento AS. Tetrazoles as PPARγ ligands: A structural and computational investigation. J Mol Graph Model 2021; 106:107932. [PMID: 33946041 DOI: 10.1016/j.jmgm.2021.107932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/17/2021] [Accepted: 04/19/2021] [Indexed: 11/18/2022]
Abstract
Diabetes is an important chronic disease affecting about 10% of the adult population in the US and over 420 million people worldwide, resulting in 1.6 million deaths every year, according to the World Health Organization. The most common type of the disease, type 2 diabetes, can be pharmacologically managed using oral hypoglycemic agents or thiazolidinediones (TZDs), such as pioglitazone, which act by activating the Peroxisome Proliferated-Activated Receptor γ. Despite their beneficial effects in diabetes treatment, TZDs like rosiglitazone and troglitazone were withdrawn due to safety reasons, creating a void in the pharmacological options for the treatment of this important disease. Here, we explored a structure-based approach in the screening for new chemical probes for a deeper investigation of the effects of PPARγ activation. A class of tetrazole compounds was identified and the compounds named T1, T2 and T3 were purchased and evaluated for their ability to interact with the PPARγ ligand binding domain (LBD). The compounds were binders with micromolar range affinity, as determined by their IC50 values. A Monte Carlo simulation of the compound T2 revealed that the tetrazole ring makes favorable interaction with the polar arm of the receptor binding pocket. Finally, the crystal structure of the PPARγ-LBD-T2 complex was solved at 2.3 Å, confirming the binding mode for this compound. The structure also revealed that, when the helix H12 is mispositioned, an alternative binding conformation is observed for the ligand suggesting an H12-dependent binding conformation for the tetrazole compound.
Collapse
Affiliation(s)
- Karina de Paula
- Grupo de Biotecnologia Molecular, Instituto de Física de São Carlos, Universidade de São Paulo, São Carlos, SP, 13566-590, Brazil
| | - Jademilson C Santos
- Grupo de Biotecnologia Molecular, Instituto de Física de São Carlos, Universidade de São Paulo, São Carlos, SP, 13566-590, Brazil
| | - Ana Carolina Mafud
- Grupo de Biotecnologia Molecular, Instituto de Física de São Carlos, Universidade de São Paulo, São Carlos, SP, 13566-590, Brazil
| | - Alessandro S Nascimento
- Grupo de Biotecnologia Molecular, Instituto de Física de São Carlos, Universidade de São Paulo, São Carlos, SP, 13566-590, Brazil.
| |
Collapse
|
46
|
Maeda R, Kami D, Shikuma A, Suzuki Y, Taya T, Matoba S, Gojo S. RNA decay in processing bodies is indispensable for adipogenesis. Cell Death Dis 2021; 12:285. [PMID: 33731683 PMCID: PMC7969960 DOI: 10.1038/s41419-021-03537-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 02/16/2021] [Accepted: 02/19/2021] [Indexed: 12/19/2022]
Abstract
The RNA decay pathway plays key regulatory roles in cell identities and differentiation processes. Although adipogenesis is transcriptionally and epigenetically regulated and has been thoroughly investigated, how RNA metabolism that contributes to the stability of phenotype-shaping transcriptomes participates in differentiation remains elusive. In this study, we investigated Ddx6, an essential component of processing bodies (PBs) that executes RNA decay and translational repression in the cytoplasm and participates in the cellular transition of reprogramming. Upon adipogenic induction, Ddx6 dynamically accumulated to form PBs with a binding partner, 4E-T, at the early phase prior to emergence of intracellular lipid droplets. In contrast, preadipocytes with Ddx6 knockout (KO) or 4E-T knockdown (KD) failed to generate PBs, resulting in significant suppression of adipogenesis. Transcription factors related to preadipocytes and negative regulators of adipogenesis that were not expressed under adipogenic stimulation were maintained in Ddx6-KO and 4E-T-KD preadipocytes under adipogenic induction. Elimination of Dlk1, a major negative regulator of adipogenesis, in 3T3L1 Ddx6-KO cells did not restore adipogenic differentiation capacity to any extent. Similar to murine cells, human primary mesenchymal stem cells, which can differentiate into adipocytes upon stimulation with adipogenic cocktails, required DDX6 to maturate into adipocytes. Therefore, RNA decay of the entire parental transcriptome, rather than removal of a strong negative regulator, could be indispensable for adipogenesis.
Collapse
Affiliation(s)
- Ryotaro Maeda
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Daisuke Kami
- Department of Regenerative Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Akira Shikuma
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yosuke Suzuki
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Toshihiko Taya
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Satoshi Gojo
- Department of Regenerative Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| |
Collapse
|
47
|
Erucic Acid-Rich Yellow Mustard Oil Improves Insulin Resistance in KK-A y Mice. Molecules 2021; 26:molecules26030546. [PMID: 33494317 PMCID: PMC7864507 DOI: 10.3390/molecules26030546] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/07/2021] [Accepted: 01/16/2021] [Indexed: 12/24/2022] Open
Abstract
Obesity is a major risk factor for some metabolic disorders including type 2 diabetes. Enhancement of peroxisome proliferator-activated receptor (PPAR) γ, a master regulator of adipocyte differentiation, is known to increase insulin-sensitive small adipocytes. In contrast, decreased PPARγ activity is also reported to improve insulin resistance. We have previously identified erucic acid as a novel natural component suppressing PPARγ transcriptional activity. In this study, we investigated the effect of erucic acid-rich yellow mustard oil (YMO) on obese/diabetic KK-Ay mice. An in vitro luciferase reporter assay and mesenchymal stem cell (MSC) differentiation assay revealed that 25 µg/mL YMO significantly inhibited PPARγ transcriptional activity and differentiation of MSCs into adipocytes but promoted their differentiation into osteoblasts. In KK-Ay mice, dietary intake of 7.0% (w/w) YMO significantly decreased the surrogate indexes for insulin resistance and the infiltration of macrophages into adipose tissue. Furthermore, 7.0% YMO increased bone mineral density. These results suggest that YMO can ameliorate obesity-induced metabolic disorders.
Collapse
|
48
|
Caioni G, Viscido A, d’Angelo M, Panella G, Castelli V, Merola C, Frieri G, Latella G, Cimini A, Benedetti E. Inflammatory Bowel Disease: New Insights into the Interplay between Environmental Factors and PPARγ. Int J Mol Sci 2021; 22:985. [PMID: 33498177 PMCID: PMC7863964 DOI: 10.3390/ijms22030985] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/14/2021] [Accepted: 01/18/2021] [Indexed: 12/19/2022] Open
Abstract
The pathophysiological processes of inflammatory bowel diseases (IBDs), i.e., Crohn's disease (CD) and ulcerative colitis (UC), are still not completely understood. The exact etiology remains unknown, but it is well established that the pathogenesis of the inflammatory lesions is due to a dysregulation of the gut immune system resulting in over-production of pro-inflammatory cytokines. Increasing evidence underlines the involvement of both environmental and genetic factors. Regarding the environment, the microbiota seems to play a crucial role. Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors that exert pleiotropic effects on glucose homeostasis, lipid metabolism, inflammatory/immune processes, cell proliferation, and fibrosis. Furthermore, PPARs modulate interactions with several environmental factors, including microbiota. A significantly impaired PPARγ expression was observed in UC patients' colonic epithelial cells, suggesting that the disruption of PPARγ signaling may represent a critical step of the IBD pathogenesis. This paper will focus on the role of PPARγ in the interaction between environmental factors and IBD, and it will analyze the most suitable in vitro and in vivo models available to better study these relationships.
Collapse
Affiliation(s)
- Giulia Caioni
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.C.); (A.V.); (M.d.); (G.P.); (V.C.); (G.F.); (G.L.); (A.C.)
| | - Angelo Viscido
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.C.); (A.V.); (M.d.); (G.P.); (V.C.); (G.F.); (G.L.); (A.C.)
| | - Michele d’Angelo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.C.); (A.V.); (M.d.); (G.P.); (V.C.); (G.F.); (G.L.); (A.C.)
| | - Gloria Panella
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.C.); (A.V.); (M.d.); (G.P.); (V.C.); (G.F.); (G.L.); (A.C.)
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Via Balzarini 1, 64100 Teramo, Italy;
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.C.); (A.V.); (M.d.); (G.P.); (V.C.); (G.F.); (G.L.); (A.C.)
| | - Carmine Merola
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Via Balzarini 1, 64100 Teramo, Italy;
| | - Giuseppe Frieri
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.C.); (A.V.); (M.d.); (G.P.); (V.C.); (G.F.); (G.L.); (A.C.)
| | - Giovanni Latella
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.C.); (A.V.); (M.d.); (G.P.); (V.C.); (G.F.); (G.L.); (A.C.)
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.C.); (A.V.); (M.d.); (G.P.); (V.C.); (G.F.); (G.L.); (A.C.)
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, Philadelphia, PA 19122, USA
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.C.); (A.V.); (M.d.); (G.P.); (V.C.); (G.F.); (G.L.); (A.C.)
| |
Collapse
|
49
|
Al-Jaber H, Al-Mansoori L, Elrayess MA. GATA-3 as a Potential Therapeutic Target for Insulin Resistance and Type 2 Diabetes Mellitus. Curr Diabetes Rev 2021; 17:169-179. [PMID: 32628587 DOI: 10.2174/1573399816666200705210417] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/09/2020] [Accepted: 06/11/2020] [Indexed: 11/22/2022]
Abstract
Impaired adipogenesis plays an important role in the development of obesity-associated insulin resistance and type 2 diabetes as it leads to ectopic fat deposition. The anti-adipogenic transcription factor GATA-3 was identified as one of the potential molecular targets responsible for the impairment of adipogenesis. The expression of GATA-3 is higher in insulinresistant obese individuals compared to BMI-matched insulin-sensitive counterparts. Adipose tissue inflammation is a crucial mediator of this process. Hyperglycemia mediates the activation of the immune system, partially through upregulation of GATA- 3, causing exacerbation of the inflammatory state associated with obesity. This review discusses the evidence supporting the inhibition of GATA-3 as a useful therapeutic strategy in obesity-associated insulin resistance and type 2 diabetes, through up-regulation adipogenesis and amelioration of the immune response.
Collapse
Affiliation(s)
- Hend Al-Jaber
- Biomedical Research Center, Qatar University, Doha, Qatar
| | | | | |
Collapse
|
50
|
Lim K, Haider A, Adams C, Sleigh A, Savage DB. Lipodistrophy: a paradigm for understanding the consequences of "overloading" adipose tissue. Physiol Rev 2020; 101:907-993. [PMID: 33356916 DOI: 10.1152/physrev.00032.2020] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Lipodystrophies have been recognized since at least the nineteenth century and, despite their rarity, tended to attract considerable medical attention because of the severity and somewhat paradoxical nature of the associated metabolic disease that so closely mimics that of obesity. Within the last 20 yr most of the monogenic subtypes have been characterized, facilitating family genetic screening and earlier disease detection as well as providing important insights into adipocyte biology and the systemic consequences of impaired adipocyte function. Even more recently, compelling genetic studies have suggested that subtle partial lipodystrophy is likely to be a major factor in prevalent insulin-resistant type 2 diabetes mellitus (T2DM), justifying the longstanding interest in these disorders. This progress has also underpinned novel approaches to treatment that, in at least some patients, can be of considerable therapeutic benefit.
Collapse
Affiliation(s)
- Koini Lim
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Afreen Haider
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Claire Adams
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Alison Sleigh
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - David B Savage
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|