1
|
Liu T, Liu Y, Shi H, Yue G, Dai X, Zhang Y, Wang S, Shen J, Xu T, Yin J, Zhang J, Zhang Y, Wang L, Zhou G, Zhang D. Sijunzi decoction improves lipid metabolism via regulation of Wnt/β-catenin signaling pathway in diabetic mice and 3T3-L1 cells. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119672. [PMID: 40139576 DOI: 10.1016/j.jep.2025.119672] [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: 09/30/2024] [Revised: 12/20/2024] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sijunzi decoction (SJZD), a traditional Chinese medicinal formula with the functions of invigorating the Spleen and replenishing Qi, has been clinically used for the management of diabetes, but its actions and underlying mechanisms on diabetic lipid metabolism remain largely unknown. AIM OF THE STUDY To explore the effects of SJZD on lipid metabolism disorders and its association with the Wnt/β-catenin pathway in the white adipose tissue of diabetic mice and 3T3-L1 cells. MATERIALS AND METHODS The diabetic lipid metabolism disorders models were established by high-fat diet/streptozotocin in mice and palmitic acid in 3T3-L1 cells, respectively. The effects of SJZD on total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL), high-density lipoprotein cholesterol (HDL), and glucose consumption were determined by biochemical assay. Hematoxylin-eosin (H&E) staining was used to examine pathological alterations in adipose tissues. Oil red O staining was used to evaluate the differentiation of lipid droplets in 3T3-L1 adipocytes. The expression levels of Wnt10b, β-catenin, CCAAT enhancer binding protein α (C/EBP-α), sterol regulatory element-binding protein 1 (SREBP-1c), acetyl-CoA carboxylase-1 (ACC1), peroxisome proliferator-activated receptor γ (PPAR-γ), fatty acid synthase (FASN), adipose triglyceride lipase (ATGL) were examined by Western blot and/or qRT-PCR. In addition, the main components of SJZD and SJZD-containing serum (SCS) were identified by UPLC-Q-TOF-MS/MS. RESULTS SJZD reduces the body fat ratio, fasting blood glucose levels, adipose tissue index and serum levels of TC, TG, and LDL, and increases the body weight, lean ratio, and serum HDL levels, as well as prevents the adipocytes hypertrophy in diabetic mice. In addition, SJZD, SCS and its ingredients (Ginsenoside Rb1 and Glycyrrhetinic acid) inhibit lipid synthesis, TG levels and promote glucose consumption in diabetic mice and 3T3-L1 adipocytes. These interventions decrease the expressions of PPAR-γ, SREBP-1c, C/EBP-α, FASN, ACC1 and P-β-catenin, and increase the expressions of ATGL, Wnt10b and β-catenin. CONCLUSION SJZD regulates the Wnt/β-catenin signaling pathway to preserve lipid metabolism homeostasis in diabetes. GRb and Gac may constitute the material basis of SJZD in attenuation of diabetic lipid metabolism disorders. These findings highlight a novel strategy for the treatment of diabetes.
Collapse
Affiliation(s)
- Tianyuan Liu
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Yage Liu
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China; Food and Pharmacy College, Xuchang University, Xuchang, Henan, 461000, PR China
| | - Hanfen Shi
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Gaiyue Yue
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Xuan Dai
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Yueyi Zhang
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Shan Wang
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Jin Shen
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Tianshu Xu
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Jiyuan Yin
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Jianglan Zhang
- Department of Pathology, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, PR China
| | - Yanfei Zhang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, PR China
| | - Lili Wang
- Department of TCM Pharmacology, Chinese Material Medica School, Beijing University of Chinese Medicine, Beijing, 102488, PR China
| | - Gang Zhou
- Center for Drug Evaluation, National Medical Products Administration, Beijing, 100076, PR China
| | - Dongwei Zhang
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China.
| |
Collapse
|
2
|
Guo G, Wang W, Tu M, Zhao B, Han J, Li J, Pan Y, Zhou J, Ma W, Liu Y, Sun T, Han X, An Y. Deciphering adipose development: Function, differentiation and regulation. Dev Dyn 2024; 253:956-997. [PMID: 38516819 DOI: 10.1002/dvdy.708] [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: 11/07/2023] [Revised: 03/02/2024] [Accepted: 03/10/2024] [Indexed: 03/23/2024] Open
Abstract
The overdevelopment of adipose tissues, accompanied by excess lipid accumulation and energy storage, leads to adipose deposition and obesity. With the increasing incidence of obesity in recent years, obesity is becoming a major risk factor for human health, causing various relevant diseases (including hypertension, diabetes, osteoarthritis and cancers). Therefore, it is of significance to antagonize obesity to reduce the risk of obesity-related diseases. Excess lipid accumulation in adipose tissues is mediated by adipocyte hypertrophy (expansion of pre-existing adipocytes) or hyperplasia (increase of newly-formed adipocytes). It is necessary to prevent excessive accumulation of adipose tissues by controlling adipose development. Adipogenesis is exquisitely regulated by many factors in vivo and in vitro, including hormones, cytokines, gender and dietary components. The present review has concluded a comprehensive understanding of adipose development including its origin, classification, distribution, function, differentiation and molecular mechanisms underlying adipogenesis, which may provide potential therapeutic strategies for harnessing obesity without impairing adipose tissue function.
Collapse
Affiliation(s)
- Ge Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Wanli Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Mengjie Tu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Binbin Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jiayang Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jiali Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Yanbing Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jie Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Wen Ma
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Yi Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Tiantian Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Xu Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Yang An
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| |
Collapse
|
3
|
Meng L, Yang Y, He S, Chen H, Zhan Y, Yang R, Li Z, Zhu J, Zhou J, Li Y, Xie L, Chen G, Zheng S, Yao X, Dong R. Single-cell sequencing of the vermiform appendix during development identifies transcriptional relationships with appendicitis in preschool children. BMC Med 2024; 22:383. [PMID: 39267041 PMCID: PMC11395239 DOI: 10.1186/s12916-024-03611-9] [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: 05/24/2024] [Accepted: 09/04/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND The development of the human vermiform appendix at the cellular level, as well as its function, is not well understood. Appendicitis in preschool children, although uncommon, is associated with a high perforation rate and increased morbidity. METHODS We performed single-cell RNA sequencing (scRNA-seq) on the human appendix during fetal and pediatric stages as well as preschool-age inflammatory appendices. Transcriptional features of each cell compartment were discussed in the developing appendix. Cellular interactions and differentiation trajectories were also investigated. We compared scRNA-seq profiles from preschool appendicitis to those of matched healthy controls to reveal disease-associated changes. Bulk transcriptomic data, immunohistochemistry, and real-time quantitative PCR were used to validate the findings. RESULTS Our analysis identified 76 cell types in total and described the cellular atlas of the developing appendix. We discovered the potential role of the BMP signaling pathway in appendiceal epithelium development and identified HOXC8 and PITX2 as the specific regulons of appendix goblet cells. Higher pericyte coverage, endothelial angiogenesis, and goblet mucus scores together with lower epithelial and endothelial tight junction scores were found in the preschool appendix, which possibly contribute to the clinical features of preschool appendicitis. Preschool appendicitis scRNA-seq profiles revealed that the interleukin-17 signaling pathway may participate in the inflammation process. CONCLUSIONS Our study provides new insights into the development of the appendix and deepens the understanding of appendicitis in preschool children.
Collapse
Affiliation(s)
- Lingdu Meng
- Department of Pediatric Surgery, Shanghai Key Laboratory of Birth Defect, Children's Hospital of Fudan University, 399 Wan Yuan Road, Shanghai, 201102, China
| | - Yifan Yang
- Department of Pediatric Surgery, Shanghai Key Laboratory of Birth Defect, Children's Hospital of Fudan University, 399 Wan Yuan Road, Shanghai, 201102, China
| | - Shiwei He
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fujian, China
| | - Huifen Chen
- Department of Pediatric Surgery, Shanghai Key Laboratory of Birth Defect, Children's Hospital of Fudan University, 399 Wan Yuan Road, Shanghai, 201102, China
| | - Yong Zhan
- Department of Pediatric Surgery, Shanghai Key Laboratory of Birth Defect, Children's Hospital of Fudan University, 399 Wan Yuan Road, Shanghai, 201102, China
| | - Ran Yang
- Department of Pediatric Surgery, Shanghai Key Laboratory of Birth Defect, Children's Hospital of Fudan University, 399 Wan Yuan Road, Shanghai, 201102, China
| | - Zifeng Li
- Department of Pediatric Surgery, Shanghai Key Laboratory of Birth Defect, Children's Hospital of Fudan University, 399 Wan Yuan Road, Shanghai, 201102, China
| | - Jiajie Zhu
- Department of Pediatric Surgery, Shanghai Key Laboratory of Birth Defect, Children's Hospital of Fudan University, 399 Wan Yuan Road, Shanghai, 201102, China
| | - Jin Zhou
- Department of Pediatric Surgery, Shanghai Key Laboratory of Birth Defect, Children's Hospital of Fudan University, 399 Wan Yuan Road, Shanghai, 201102, China
| | - Yi Li
- Department of Pediatric Surgery, Shanghai Key Laboratory of Birth Defect, Children's Hospital of Fudan University, 399 Wan Yuan Road, Shanghai, 201102, China
| | - Lulu Xie
- Department of Pediatric Surgery, Shanghai Key Laboratory of Birth Defect, Children's Hospital of Fudan University, 399 Wan Yuan Road, Shanghai, 201102, China
| | - Gong Chen
- Department of Pediatric Surgery, Shanghai Key Laboratory of Birth Defect, Children's Hospital of Fudan University, 399 Wan Yuan Road, Shanghai, 201102, China
| | - Shan Zheng
- Department of Pediatric Surgery, Shanghai Key Laboratory of Birth Defect, Children's Hospital of Fudan University, 399 Wan Yuan Road, Shanghai, 201102, China.
| | - Xiaoying Yao
- Family Planning Department, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.
| | - Rui Dong
- Department of Pediatric Surgery, Shanghai Key Laboratory of Birth Defect, Children's Hospital of Fudan University, 399 Wan Yuan Road, Shanghai, 201102, China.
| |
Collapse
|
4
|
Byun KA, Kim HM, Oh S, Batsukh S, Lee S, Oh M, Lee J, Lee R, Kim JW, Oh SM, Kim J, Kim G, Park HJ, Hong H, Lee J, An SH, Oh SS, Jung YS, Son KH, Byun K. High-Intensity Focused Ultrasound Increases Facial Adipogenesis in a Swine Model via Modulation of Adipose-Derived Stem Cell Cilia. Int J Mol Sci 2024; 25:7648. [PMID: 39062891 PMCID: PMC11277104 DOI: 10.3390/ijms25147648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Decreased medial cheek fat volume during aging leads to loss of a youthful facial shape. Increasing facial volume by methods such as adipose-derived stem cell (ASC) injection can produce facial rejuvenation. High-intensity focused ultrasound (HIFU) can increase adipogenesis in subcutaneous fat by modulating cilia on ASCs, which is accompanied by increased HSP70 and decreased NF-κB expression. Thus, we evaluated the effect of HIFU on increasing facial adipogenesis in swine (n = 2) via modulation of ASC cilia. Expression of CD166, an ASC marker, differed by subcutaneous adipose tissue location. CD166 expression in the zygomatic arch (ZA) was significantly higher than that in the subcutaneous adipose tissue in the mandible or lateral temporal areas. HIFU was applied only on the right side of the face, which was compared with the left side, where HIFU was not applied, as a control. HIFU produced a significant increase in HSP70 expression, decreased expression of NF-κB and a cilia disassembly factor (AURKA), and increased expression of a cilia increasing factor (ARL13B) and PPARG and CEBPA, which are the main regulators of adipogenesis. All of these changes were most prominent at the ZA. Facial adipose tissue thickness was also increased by HIFU. Adipose tissue volume, evaluated by magnetic resonance imaging, was increased by HIFU, most prominently in the ZA. In conclusion, HIFU increased ASC marker expression, accompanied by increased HSP70 and decreased NF-κB expression. Additionally, changes in cilia disassembly and length and expression of adipogenesis were observed. These results suggest that HIFU could be used to increase facial volume by modulating adipogenesis.
Collapse
Affiliation(s)
- Kyung-A Byun
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
- LIBON Inc., Incheon 22006, Republic of Korea
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Hyoung Moon Kim
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
- Maylin Clinic, Goyang 10391, Republic of Korea
| | - Seyeon Oh
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Sosorburam Batsukh
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Sangsu Lee
- Mirabel Clinic, Seoul 04596, Republic of Korea
| | - Myungjune Oh
- GangnamON Clinic, Seoul 06129, Republic of Korea
| | | | - Ran Lee
- Ezen Clinic, Cheonan 31090, Republic of Korea
| | - Jae Woo Kim
- Lienjang Clinic, Seoul 04536, Republic of Korea
| | - Seung Min Oh
- GangnamON Clinic, Seoul 06129, Republic of Korea
| | - Jisun Kim
- MH Clinic, Seoul 06010, Republic of Korea
| | - Geebum Kim
- Misogain Dermatology Clinic, Gimpo 10108, Republic of Korea
| | - Hyun Jun Park
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
- Maylin Clinic the Cheongdam, Seoul 06091, Republic of Korea
| | - Hanbit Hong
- Lux Well Clinic, Cheongju 28424, Republic of Korea
| | - Jehyuk Lee
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
- Doctorbom Clinic, Seoul 06614, Republic of Korea
| | - Sang-Hyun An
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu 41061, Republic of Korea
| | - Sung Suk Oh
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu 41061, Republic of Korea
| | - Yeon-Seop Jung
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu 41061, Republic of Korea
| | - Kuk Hui Son
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, Gachon University, Incheon 21565, Republic of Korea
| | - Kyunghee Byun
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health & Sciences and Technology (GAIHST), Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
5
|
Nellinger S, Kluger PJ. How Mechanical and Physicochemical Material Characteristics Influence Adipose-Derived Stem Cell Fate. Int J Mol Sci 2023; 24:ijms24043551. [PMID: 36834966 PMCID: PMC9961531 DOI: 10.3390/ijms24043551] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/28/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Adipose-derived stem cells (ASCs) are a subpopulation of mesenchymal stem cells. Compared to bone marrow-derived stem cells, they can be harvested with minimal invasiveness. ASCs can be easily expanded and were shown to be able to differentiate into several clinically relevant cell types. Therefore, this cell type represents a promising component in various tissue engineering and medical approaches (e.g., cell therapy). In vivo cells are surrounded by the extracellular matrix (ECM) that provides a wide range of tissue-specific physical and chemical cues, such as stiffness, topography, and chemical composition. Cells can sense the characteristics of their ECM and respond to them in a specific cellular behavior (e.g., proliferation or differentiation). Thus, in vitro biomaterial properties represent an important tool to control ASCs behavior. In this review, we give an overview of the current research in the mechanosensing of ASCs and current studies investigating the impact of material stiffens, topography, and chemical modification on ASC behavior. Additionally, we outline the use of natural ECM as a biomaterial and its interaction with ASCs regarding cellular behavior.
Collapse
Affiliation(s)
- Svenja Nellinger
- Reutlingen Research Institute, Reutlingen University, 72762 Reutlingen, Germany
| | - Petra Juliane Kluger
- School of Life Sciences, Reutlingen University, 72762 Reutlingen, Germany
- Correspondence: ; Tel.: +49-07121-271-2061
| |
Collapse
|
6
|
Chen B, Shi Z, Wang Y, Chen M, Yang C, Cui H, Su T, Kwan HY. Discovery of a novel anti-obesity meroterpenoid agent targeted subcutaneous adipose tissue. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 106:154396. [PMID: 36057145 DOI: 10.1016/j.phymed.2022.154396] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/28/2022] [Accepted: 08/17/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Meroterpenoid furanasperterpene A (T2-3) with a novel 6/6/6/6/5 pentacyclic skeleton was isolated from the Aspergillus terreus GZU-31-1. Previously, we showed that T2-3 possessed significant lipid-lowering effects in 3T3-L1 adipocytes at 5 μM concentration. However, its therapeutic effect in metabolic disease and the underlying mechanisms of action remain unclear. METHODS High fat diet-induced obesity (DIO) mouse model and 3T3-L1 cell model were used to assess the anti-obesity effects of T2-3. Lipids in the adipocytes were examined by Oil Red O staining. β-catenin expression was examined by immunofluorescence and Western blotting, its activity was assessed by TOPflash/FOPflash assay. RESULTS T2-3 possessed potent anti-obesity effects in DIO mice, it significantly reduced body weight and subcutaneous adipose tissue (SAT) mass. Mechanistic studies showed that T2-3 significantly inhibited 3T3-L1 preadipocyte differentiation as indicated by the reduced number of mature adipocytes. The treatments also reduced the expressions of critical adipogenic transcription factors CEBP-α and PPAR-γ in both 3T3-L1 adipocytes and SAT in DIO mice. Interestingly, T2-3 increased the cytoplasmic and nuclear expressions of β-catenin and the transcriptional activity of β-catenin in 3T3-L1 adipocytes; the elevated β-catenin expression was also observed in SAT of the T2-3-treated DIO mice. Indeed, upregulation of β-catenin activity suppressed adipogenesis, while β-catenin inhibitor JW67 reversed the anti-adipogenic effect of T2-3. Taken together, our data suggest that T2-3 inhibits adipogenesis by upregulating β-catenin activity. CONCLUSIONS Our study is the first report demonstrating meroterpenoid furanasperterpene A as a novel 6/6/6/6/5 pentacyclic skeleton (T2-3) that possesses potent anti-adipogenic effect by targeting β-catenin signaling pathway. Our findings drive new anti-obesity drug discovery and provide drug leads for chemists and pharmacologists.
Collapse
Affiliation(s)
- Baisen Chen
- Centre for Cancer & Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| | - Zhiqiang Shi
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yechun Wang
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Minting Chen
- Centre for Cancer & Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Chunfang Yang
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Hui Cui
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China; School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Tao Su
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Hiu Yee Kwan
- Centre for Cancer & Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| |
Collapse
|
7
|
Magusto J, Beaupère C, Afonso MB, Auclair M, Delaunay JL, Soret PA, Courtois G, Aït-Slimane T, Housset C, Jéru I, Fève B, Ratziu V, Rodrigues CM, Gautheron J. The necroptosis-inducing pseudokinase mixed lineage kinase domain-like regulates the adipogenic differentiation of pre-adipocytes. iScience 2022; 25:105166. [PMID: 36204273 PMCID: PMC9530846 DOI: 10.1016/j.isci.2022.105166] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/02/2022] [Accepted: 09/16/2022] [Indexed: 11/28/2022] Open
Abstract
Receptor-interacting protein kinase-3 (RIPK3) and mixed lineage kinase domain-like (MLKL) proteins are key regulators of necroptosis, a highly pro-inflammatory mode of cell death, which has been involved in various human diseases. Necroptotic-independent functions of RIPK3 and MLKL also exist, notably in the adipose tissue but remain poorly defined. Using knock-out (KO) cell models, we investigated the role of RIPK3 and MLKL in adipocyte differentiation. Mlkl-KO abolished white adipocyte differentiation via a strong expression of Wnt10b, a ligand of the Wnt/β-catenin pathway, and a downregulation of genes involved in lipid metabolism. This effect was not recapitulated by the ablation of Ripk3. Conversely, Mlkl and Ripk3 deficiencies did not block beige adipocyte differentiation. These findings indicate that RIPK3 and MLKL have distinct roles in adipogenesis. The absence of MLKL blocks the differentiation of white, but not beige, adipocytes highlighting the therapeutic potential of MLKL inhibition in obesity. Mlkl deficiency inhibits white, but not beige, adipocyte differentiation MLKL deficiency suppresses the expression of master regulators of adipogenesis Mlkl deficiency up-regulates Wnt10b expression Ripk3 deficiency does not alter white and beige adipocyte differentiation
Collapse
|
8
|
Szymczak-Pajor I, Miazek K, Selmi A, Balcerczyk A, Śliwińska A. The Action of Vitamin D in Adipose Tissue: Is There the Link between Vitamin D Deficiency and Adipose Tissue-Related Metabolic Disorders? Int J Mol Sci 2022; 23:956. [PMID: 35055140 PMCID: PMC8779075 DOI: 10.3390/ijms23020956] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/09/2022] [Accepted: 01/10/2022] [Indexed: 12/11/2022] Open
Abstract
Adipose tissue plays an important role in systemic metabolism via the secretion of adipocytokines and storing and releasing energy. In obesity, adipose tissue becomes dysfunctional and characterized by hypertrophied adipocytes, increased inflammation, hypoxia, and decreased angiogenesis. Although adipose tissue is one of the major stores of vitamin D, its deficiency is detective in obese subjects. In the presented review, we show how vitamin D regulates numerous processes in adipose tissue and how their dysregulation leads to metabolic disorders. The molecular response to vitamin D in adipose tissue affects not only energy metabolism and adipokine and anti-inflammatory cytokine production via the regulation of gene expression but also genes participating in antioxidant defense, adipocytes differentiation, and apoptosis. Thus, its deficiency disturbs adipocytokines secretion, metabolism, lipid storage, adipogenesis, thermogenesis, the regulation of inflammation, and oxidative stress balance. Restoring the proper functionality of adipose tissue in overweight or obese subjects is of particular importance in order to reduce the risk of developing obesity-related complications, such as cardiovascular diseases and diabetes. Taking into account the results of experimental studies, it seemed that vitamin D may be a remedy for adipose tissue dysfunction, but the results of the clinical trials are not consistent, as some of them show improvement and others no effect of this vitamin on metabolic and insulin resistance parameters. Therefore, further studies are required to evaluate the beneficial effects of vitamin D, especially in overweight and obese subjects, due to the presence of a volumetric dilution of this vitamin among them.
Collapse
Affiliation(s)
- Izabela Szymczak-Pajor
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland;
| | - Krystian Miazek
- Institute of Applied Radiation Chemistry, Faculty of Chemistry, Lodz University of Technology, 15 Wroblewskiego, 93-590 Lodz, Poland;
| | - Anna Selmi
- Department of Molecular Biophysics, University of Lodz, 141/143 Pomorska, 90-236 Lodz, Poland; (A.S.); (A.B.)
| | - Aneta Balcerczyk
- Department of Molecular Biophysics, University of Lodz, 141/143 Pomorska, 90-236 Lodz, Poland; (A.S.); (A.B.)
| | - Agnieszka Śliwińska
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland;
| |
Collapse
|
9
|
Briganti S, Truglio M, Angiolillo A, Lombardo S, Leccese D, Camera E, Picardo M, Di Costanzo A. Application of Sebum Lipidomics to Biomarkers Discovery in Neurodegenerative Diseases. Metabolites 2021; 11:metabo11120819. [PMID: 34940576 PMCID: PMC8708591 DOI: 10.3390/metabo11120819] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/16/2021] [Accepted: 11/24/2021] [Indexed: 02/06/2023] Open
Abstract
Lipidomics is strategic in the discovery of biomarkers of neurodegenerative diseases (NDDs). The skin surface lipidome bears the potential to provide biomarker candidates in the detection of pathological processes occurring in distal organs. We investigated the sebum composition to search diagnostic and, possibly, prognostic, biomarkers of Alzheimer’s disease (AD) and Parkinson’s disease (PD). The observational study included 64 subjects: 20 characterized as “probable AD with documented decline”, 20 as “clinically established PD”, and 24 healthy subjects (HS) of comparable age. The analysis of sebum by GCMS and TLC retrieved the amounts (µg) of 41 free fatty acids (FFAs), 7 fatty alcohols (FOHs), vitamin E, cholesterol, squalene, and total triglycerides (TGs) and wax esters (WEs). Distributions of sebum lipids in NDDs and healthy conditions were investigated with multivariate ANOVA-simultaneous component analysis (ASCA). The deranged sebum composition associated with the PD group showed incretion of most composing lipids compared to HS, whereas only two lipid species (vitamin E and FOH14:0) were discriminant of AD samples and presented lower levels than HS sebum. Thus, sebum lipid biosynthetic pathways are differently affected in PD and AD. The characteristic sebum bio-signatures detected support the value of sebum lipidomics in the biomarkers search in NDDs.
Collapse
Affiliation(s)
- Stefania Briganti
- Laboratory of Cutaneous Physiopathology, San Gallicano Dermatological Institute—IRCCS, Via Elio Chianesi 53, 00144 Rome, Italy; (S.B.); (M.T.); (M.P.)
| | - Mauro Truglio
- Laboratory of Cutaneous Physiopathology, San Gallicano Dermatological Institute—IRCCS, Via Elio Chianesi 53, 00144 Rome, Italy; (S.B.); (M.T.); (M.P.)
| | - Antonella Angiolillo
- Centre for Research and Training in Medicine of Aging, Department of Medicine and Health Science “V. Tiberio”, University of Molise, Via De Santis, 86100 Campobasso, Italy; (A.A.); (S.L.); (D.L.); (A.D.C.)
| | - Salvatore Lombardo
- Centre for Research and Training in Medicine of Aging, Department of Medicine and Health Science “V. Tiberio”, University of Molise, Via De Santis, 86100 Campobasso, Italy; (A.A.); (S.L.); (D.L.); (A.D.C.)
| | - Deborah Leccese
- Centre for Research and Training in Medicine of Aging, Department of Medicine and Health Science “V. Tiberio”, University of Molise, Via De Santis, 86100 Campobasso, Italy; (A.A.); (S.L.); (D.L.); (A.D.C.)
| | - Emanuela Camera
- Laboratory of Cutaneous Physiopathology, San Gallicano Dermatological Institute—IRCCS, Via Elio Chianesi 53, 00144 Rome, Italy; (S.B.); (M.T.); (M.P.)
- Correspondence: ; Tel.: +39-06-5266-6241; Fax: +39-06-5266-6247
| | - Mauro Picardo
- Laboratory of Cutaneous Physiopathology, San Gallicano Dermatological Institute—IRCCS, Via Elio Chianesi 53, 00144 Rome, Italy; (S.B.); (M.T.); (M.P.)
| | - Alfonso Di Costanzo
- Centre for Research and Training in Medicine of Aging, Department of Medicine and Health Science “V. Tiberio”, University of Molise, Via De Santis, 86100 Campobasso, Italy; (A.A.); (S.L.); (D.L.); (A.D.C.)
| |
Collapse
|
10
|
Dutta P, Kodigepalli KM, LaHaye S, Thompson JW, Rains S, Nagel C, Thatcher K, Hinton RB, Lincoln J. KPT-330 Prevents Aortic Valve Calcification via a Novel C/EBPβ Signaling Pathway. Circ Res 2021; 128:1300-1316. [PMID: 33601919 PMCID: PMC8085092 DOI: 10.1161/circresaha.120.318503] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Punashi Dutta
- Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
- Pediatric Cardiology, The Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI, USA
| | - Karthik M. Kodigepalli
- Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
- Pediatric Cardiology, The Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI, USA
| | - Stephanie LaHaye
- The Institute for Genomic Medicine at Nationwide Children’s Hospital, Columbus, OH, USA
| | - J. Will Thompson
- Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Sarah Rains
- Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
- Duke Proteomics and Metabolomics Shared Resource, Durham, NC, USA
| | - Casey Nagel
- Ocean Ridge Biosciences, Deerfield Beach, Florida, USA
| | - Kaitlyn Thatcher
- Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
- Pediatric Cardiology, The Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI, USA
| | - Robert B. Hinton
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Joy Lincoln
- Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
- Pediatric Cardiology, The Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
11
|
Abstract
Aortic stenosis (AS) remains one of the most common forms of valve disease, with significant impact on patient survival. The disease is characterized by left ventricular outflow obstruction and encompasses a series of stenotic lesions starting from the left ventricular outflow tract to the descending aorta. Obstructions may be subvalvar, valvar, or supravalvar and can be present at birth (congenital) or acquired later in life. Bicuspid aortic valve, whereby the aortic valve forms with two instead of three cusps, is the most common cause of AS in younger patients due to primary anatomic narrowing of the valve. In addition, the secondary onset of premature calcification, likely induced by altered hemodynamics, further obstructs left ventricular outflow in bicuspid aortic valve patients. In adults, degenerative AS involves progressive calcification of an anatomically normal, tricuspid aortic valve and is attributed to lifelong exposure to multifactoral risk factors and physiological wear-and-tear that negatively impacts valve structure-function relationships. AS continues to be the most frequent valvular disease that requires intervention, and aortic valve replacement is the standard treatment for patients with severe or symptomatic AS. While the positive impacts of surgical interventions are well documented, the financial burden, the potential need for repeated procedures, and operative risks are substantial. In addition, the clinical management of asymptomatic patients remains controversial. Therefore, there is a critical need to develop alternative approaches to prevent the progression of left ventricular outflow obstruction, especially in valvar lesions. This review summarizes our current understandings of AS cause; beginning with developmental origins of congenital valve disease, and leading into the multifactorial nature of AS in the adult population.
Collapse
Affiliation(s)
- Punashi Dutta
- The Herma Heart Institute, Section of Pediatric Cardiology, Children's Wisconsin, Milwaukee, WI (P.D., J.F.J., H.K., J.L.).,Department of Pediatrics, Medical College of Wisconsin, Milwaukee (P.D., J.F.J., J.L.)
| | - Jeanne F James
- The Herma Heart Institute, Section of Pediatric Cardiology, Children's Wisconsin, Milwaukee, WI (P.D., J.F.J., H.K., J.L.).,Department of Pediatrics, Medical College of Wisconsin, Milwaukee (P.D., J.F.J., J.L.)
| | - Hail Kazik
- The Herma Heart Institute, Section of Pediatric Cardiology, Children's Wisconsin, Milwaukee, WI (P.D., J.F.J., H.K., J.L.).,Department of Biomedical Engineering, Marquette University & Medical College of Wisconsin, Milwaukee (H.K.)
| | - Joy Lincoln
- The Herma Heart Institute, Section of Pediatric Cardiology, Children's Wisconsin, Milwaukee, WI (P.D., J.F.J., H.K., J.L.).,Department of Pediatrics, Medical College of Wisconsin, Milwaukee (P.D., J.F.J., J.L.)
| |
Collapse
|
12
|
Stypulkowski E, Feng Q, Joseph I, Farrell V, Flores J, Yu S, Sakamori R, Sun J, Bandyopadhyay S, Das S, Dobrowolski R, Bonder EM, Chen MH, Gao N. Rab8 attenuates Wnt signaling and is required for mesenchymal differentiation into adipocytes. J Biol Chem 2021; 296:100488. [PMID: 33662399 PMCID: PMC8042397 DOI: 10.1016/j.jbc.2021.100488] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/18/2021] [Accepted: 02/26/2021] [Indexed: 12/11/2022] Open
Abstract
Differentiation of mesenchymal stem cells into adipocyte requires coordination of external stimuli and depends upon the functionality of the primary cilium. The Rab8 small GTPases are regulators of intracellular transport of membrane-bound structural and signaling cargo. However, the physiological contribution of the intrinsic trafficking network controlled by Rab8 to mesenchymal tissue differentiation has not been fully defined in vivo and in primary tissue cultures. Here, we show that mouse embryonic fibroblasts (MEFs) lacking Rab8 have severely impaired adipocyte differentiation in vivo and ex vivo. Immunofluorescent localization and biochemical analyses of Rab8a-deficient, Rab8b-deficient, and Rab8a and Rab8b double-deficient MEFs revealed that Rab8 controls the Lrp6 vesicular compartment, clearance of basal signalosome, traffic of frizzled two receptor, and thereby a proper attenuation of Wnt signaling in differentiating MEFs. Upon induction of adipogenesis program, Rab8a- and Rab8b-deficient MEFs exhibited severely defective lipid-droplet formation and abnormal cilia morphology, despite overall intact cilia growth and ciliary cargo transport. Our results suggest that intracellular Rab8 traffic regulates induction of adipogenesis via proper positioning of Wnt receptors for signaling control in mesenchymal cells.
Collapse
Affiliation(s)
- Ewa Stypulkowski
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Qiang Feng
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Ivor Joseph
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Victoria Farrell
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Juan Flores
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Shiyan Yu
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Ryotaro Sakamori
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Jiaxin Sun
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | | | - Soumyashree Das
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Radek Dobrowolski
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Edward M Bonder
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Miao-Hsueh Chen
- Department of Pediatrics, Baylor College of Medicine, Children's Nutrition Research Center, Houston, Texas, USA.
| | - Nan Gao
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA; Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA.
| |
Collapse
|
13
|
Jakab J, Miškić B, Mikšić Š, Juranić B, Ćosić V, Schwarz D, Včev A. Adipogenesis as a Potential Anti-Obesity Target: A Review of Pharmacological Treatment and Natural Products. Diabetes Metab Syndr Obes 2021; 14:67-83. [PMID: 33447066 PMCID: PMC7802907 DOI: 10.2147/dmso.s281186] [Citation(s) in RCA: 148] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 12/09/2020] [Indexed: 12/14/2022] Open
Abstract
Obesity is recognized as a severe threat to overall human health and is associated with type 2 diabetes mellitus, dyslipidemia, hypertension, and cardiovascular diseases. Abnormal expansion of white adipose tissue involves increasing the existing adipocytes' cell size or increasing the number through the differentiation of new adipocytes. Adipogenesis is a process of proliferation and differentiation of adipocyte precursor cells in mature adipocytes. As a key process in determining the number of adipocytes, it is a possible therapeutic approach for obesity. Therefore, it is necessary to identify the molecular mechanisms involved in adipogenesis that could serve as suitable therapeutic targets. Reducing bodyweight is regarded as a major health benefit. Limited efficacy and possible side effects and drug interactions of available anti-obesity treatment highlight a constant need for finding novel efficient and safe anti-obesity ingredients. Numerous studies have recently investigated the inhibitory effects of natural products on adipocyte differentiation and lipid accumulation. Possible anti-obesity effects of natural products include the induction of apoptosis, cell-cycle arrest or delayed progression, and interference with transcription factor cascade or intracellular signaling pathways during the early phase of adipogenesis.
Collapse
Affiliation(s)
- Jelena Jakab
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
- Correspondence: Jelena Jakab Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Crkvena 21, Osijek31 000, CroatiaTel +385 91 224 1502 Email
| | - Blaženka Miškić
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
- Department of Internal Medicine, General Hospital “Dr. Josip Benčević”, Slavonski Brod, Croatia
| | - Štefica Mikšić
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Brankica Juranić
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
- Department of Cardiology, University Hospital Osijek, Osijek, Croatia
| | - Vesna Ćosić
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Dragan Schwarz
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
- Special Hospital Radiochirurgia Zagreb, Zagreb, Croatia
| | - Aleksandar Včev
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| |
Collapse
|
14
|
Han S, Kim J, Lee G, Kim D. Mechanical Properties of Materials for Stem Cell Differentiation. ACTA ACUST UNITED AC 2020; 4:e2000247. [DOI: 10.1002/adbi.202000247] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/28/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Seong‐Beom Han
- KU‐KIST Graduate School of Converging Science and Technology Korea University 145, Anam‐ro, Seongbuk‐gu Seoul 02841 Republic of Korea
| | - Jeong‐Ki Kim
- KU‐KIST Graduate School of Converging Science and Technology Korea University 145, Anam‐ro, Seongbuk‐gu Seoul 02841 Republic of Korea
| | - Geonhui Lee
- KU‐KIST Graduate School of Converging Science and Technology Korea University 145, Anam‐ro, Seongbuk‐gu Seoul 02841 Republic of Korea
| | - Dong‐Hwee Kim
- KU‐KIST Graduate School of Converging Science and Technology Korea University 145, Anam‐ro, Seongbuk‐gu Seoul 02841 Republic of Korea
| |
Collapse
|
15
|
Fan L, Xu H, Li D, Li H, Lu D. A novel long noncoding RNA, AC092834.1, regulates the adipogenic differentiation of human adipose-derived mesenchymal stem cells via the DKK1/Wnt/β-catenin signaling pathway. Biochem Biophys Res Commun 2020; 525:747-754. [PMID: 32145912 DOI: 10.1016/j.bbrc.2020.02.140] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 02/22/2020] [Indexed: 01/15/2023]
Abstract
Long noncoding RNAs (lncRNAs) have been implicated in a range of developmental processes and diseases, but the roles and mechanisms by which they act in adipogenic differentiation and adipose tissue biology are still unknown. By comparing the different expression patterns of lncRNAs before and after the adipocyte differentiation of human adipose-derived mesenchymal stem cells (hADSCs), we characterized a novel lncRNA, AC092834.1, which is significantly increased in preadipocytes. By gain- and loss-of-function experiments, we demonstrated that lncRNA AC092834.1 potentiated adipogenic differentiation through directly increasing the level of expression of DKK1, which competitively binds with LRP5 to inhibit the Wnt-β-catenin pathway and reduce the inhibition of adipogenesis by Wnt signaling. This finding provides novel mechanistic insights into a critical role for lncRNA AC092834.1 as a regulator of adipogenic differentiation, which expands our knowledge about the molecular mechanisms of obesity and other adipogenic differentiation-related disorders.
Collapse
Affiliation(s)
- Linyuan Fan
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China
| | - Haoying Xu
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No. BZO381), Beijing, 100005, China
| | - Di Li
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No. BZO381), Beijing, 100005, China
| | - Hongling Li
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No. BZO381), Beijing, 100005, China
| | - Dan Lu
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China.
| |
Collapse
|
16
|
Clayton RW, Langan EA, Ansell DM, de Vos IJHM, Göbel K, Schneider MR, Picardo M, Lim X, van Steensel MAM, Paus R. Neuroendocrinology and neurobiology of sebaceous glands. Biol Rev Camb Philos Soc 2020; 95:592-624. [PMID: 31970855 DOI: 10.1111/brv.12579] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 12/17/2019] [Accepted: 12/19/2019] [Indexed: 12/11/2022]
Abstract
The nervous system communicates with peripheral tissues through nerve fibres and the systemic release of hypothalamic and pituitary neurohormones. Communication between the nervous system and the largest human organ, skin, has traditionally received little attention. In particular, the neuro-regulation of sebaceous glands (SGs), a major skin appendage, is rarely considered. Yet, it is clear that the SG is under stringent pituitary control, and forms a fascinating, clinically relevant peripheral target organ in which to study the neuroendocrine and neural regulation of epithelia. Sebum, the major secretory product of the SG, is composed of a complex mixture of lipids resulting from the holocrine secretion of specialised epithelial cells (sebocytes). It is indicative of a role of the neuroendocrine system in SG function that excess circulating levels of growth hormone, thyroxine or prolactin result in increased sebum production (seborrhoea). Conversely, growth hormone deficiency, hypothyroidism, and adrenal insufficiency result in reduced sebum production and dry skin. Furthermore, the androgen sensitivity of SGs appears to be under neuroendocrine control, as hypophysectomy (removal of the pituitary) renders SGs largely insensitive to stimulation by testosterone, which is crucial for maintaining SG homeostasis. However, several neurohormones, such as adrenocorticotropic hormone and α-melanocyte-stimulating hormone, can stimulate sebum production independently of either the testes or the adrenal glands, further underscoring the importance of neuroendocrine control in SG biology. Moreover, sebocytes synthesise several neurohormones and express their receptors, suggestive of the presence of neuro-autocrine mechanisms of sebocyte modulation. Aside from the neuroendocrine system, it is conceivable that secretion of neuropeptides and neurotransmitters from cutaneous nerve endings may also act on sebocytes or their progenitors, given that the skin is richly innervated. However, to date, the neural controls of SG development and function remain poorly investigated and incompletely understood. Botulinum toxin-mediated or facial paresis-associated reduction of human sebum secretion suggests that cutaneous nerve-derived substances modulate lipid and inflammatory cytokine synthesis by sebocytes, possibly implicating the nervous system in acne pathogenesis. Additionally, evidence suggests that cutaneous denervation in mice alters the expression of key regulators of SG homeostasis. In this review, we examine the current evidence regarding neuroendocrine and neurobiological regulation of human SG function in physiology and pathology. We further call attention to this line of research as an instructive model for probing and therapeutically manipulating the mechanistic links between the nervous system and mammalian skin.
Collapse
Affiliation(s)
- Richard W Clayton
- Centre for Dermatology, School of Biological Sciences, University of Manchester, and NIHR Manchester Biomedical Research Centre, Stopford Building, Oxford Road, Manchester, M13 9PT, U.K.,Skin Research Institute of Singapore, Agency for Science, Technology and Research, 11 Mandalay Road, #17-01 Clinical Sciences Building, 308232, Singapore
| | - Ewan A Langan
- Centre for Dermatology, School of Biological Sciences, University of Manchester, and NIHR Manchester Biomedical Research Centre, Stopford Building, Oxford Road, Manchester, M13 9PT, U.K.,Department of Dermatology, Allergology und Venereology, University of Lübeck, Ratzeburger Allee 160, Lübeck, 23538, Germany
| | - David M Ansell
- Centre for Dermatology, School of Biological Sciences, University of Manchester, and NIHR Manchester Biomedical Research Centre, Stopford Building, Oxford Road, Manchester, M13 9PT, U.K.,Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, U.K
| | - Ivo J H M de Vos
- Skin Research Institute of Singapore, Agency for Science, Technology and Research, 11 Mandalay Road, #17-01 Clinical Sciences Building, 308232, Singapore
| | - Klaus Göbel
- Skin Research Institute of Singapore, Agency for Science, Technology and Research, 11 Mandalay Road, #17-01 Clinical Sciences Building, 308232, Singapore.,Department of Dermatology, Cologne Excellence Cluster on Stress Responses in Aging Associated Diseases (CECAD), and Centre for Molecular Medicine Cologne, The University of Cologne, Joseph-Stelzmann-Straße 26, Cologne, 50931, Germany
| | - Marlon R Schneider
- German Federal Institute for Risk Assessment (BfR), German Centre for the Protection of Laboratory Animals (Bf3R), Max-Dohrn-Straße 8-10, Berlin, 10589, Germany
| | - Mauro Picardo
- Cutaneous Physiopathology and Integrated Centre of Metabolomics Research, San Gallicano Dermatological Institute IRCCS, Via Elio Chianesi 53, Rome, 00144, Italy
| | - Xinhong Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore
| | - Maurice A M van Steensel
- Skin Research Institute of Singapore, Agency for Science, Technology and Research, 11 Mandalay Road, #17-01 Clinical Sciences Building, 308232, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore
| | - Ralf Paus
- Centre for Dermatology, School of Biological Sciences, University of Manchester, and NIHR Manchester Biomedical Research Centre, Stopford Building, Oxford Road, Manchester, M13 9PT, U.K.,Dr. Phllip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, 1600 NW 10th Avenue, RMSB 2023A, Miami, FL, 33136, U.S.A.,Monasterium Laboratory, Mendelstraße 17, Münster, 48149, Germany
| |
Collapse
|
17
|
Im DU, Kim SC, Chau GC, Um SH. Carbamazepine Enhances Adipogenesis by Inhibiting Wnt/β-catenin Expression. Cells 2019; 8:cells8111460. [PMID: 31752244 PMCID: PMC6912407 DOI: 10.3390/cells8111460] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/08/2019] [Accepted: 11/14/2019] [Indexed: 12/17/2022] Open
Abstract
Carbamazepine is a drug that is widely used in the treatment of epilepsy and bipolar disorder. The prevalence of obesity in patients treated with carbamazepine has been frequently reported. However, whether carbamazepine affects adipogenesis, one of the critical steps in the development of obesity, remains unclear. Here, we show that carbamazepine increased the expression levels of peroxisome proliferator-activated receptor γ (PPARγ), CCAAT/enhancer-binding protein β (C/EBPβ), and fatty acid synthase (FASN) in 3T3-L1 cells. Notably, carbamazepine inhibited the expression levels of β-catenin, a negative regulator of adipogenesis, leading to enhanced adipogenesis. Conversely, β-catenin overexpression abolished the effect of carbamazepine on adipogenic gene expression. However, depletion of β-catenin further enhanced PPARγ expression. In addition, carbamazepine reduced β-catenin expression by lowering the levels of phospho-low density lipoprotein receptor-related protein 6 (p-LRP6) and phospho-glycogen synthase kinase 3β (p-GSK3β) in Wnt/β-catenin signaling. Moreover, carbamazepine reduced Wnt mRNA expression and decreased the promoter activities of TCF, the target of β-catenin during adipogenesis. These results suggest that carbamazepine enhances adipogenesis by suppressing Wnt/β-catenin expression, indicating its potential effects on obesity-related metabolism.
Collapse
Affiliation(s)
- Dong Uk Im
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi-do 16419, Korea; (D.U.I.); (G.C.C.)
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Samsung Medical Center, Sungkyunkwan University, Seoul 06351, Korea;
| | - Sang Chon Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Samsung Medical Center, Sungkyunkwan University, Seoul 06351, Korea;
| | - Gia Cac Chau
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi-do 16419, Korea; (D.U.I.); (G.C.C.)
| | - Sung Hee Um
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi-do 16419, Korea; (D.U.I.); (G.C.C.)
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Samsung Medical Center, Sungkyunkwan University, Seoul 06351, Korea;
- Biomedical Institute Convergence at Sungkyunkwan University, Suwon, Gyeonggi-do 16419, Korea
- Correspondence: ; Tel.: +82-31-299-6123; Fax: +82-31-299-6109
| |
Collapse
|
18
|
Johansson J, Naszai M, Hodder MC, Pickering KA, Miller BW, Ridgway RA, Yu Y, Peschard P, Brachmann S, Campbell AD, Cordero JB, Sansom OJ. RAL GTPases Drive Intestinal Stem Cell Function and Regeneration through Internalization of WNT Signalosomes. Cell Stem Cell 2019; 24:592-607.e7. [PMID: 30853556 PMCID: PMC6459002 DOI: 10.1016/j.stem.2019.02.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 12/24/2018] [Accepted: 02/05/2019] [Indexed: 01/05/2023]
Abstract
Ral GTPases are RAS effector molecules and by implication a potential therapeutic target for RAS mutant cancer. However, very little is known about their roles in stem cells and tissue homeostasis. Using Drosophila, we identified expression of RalA in intestinal stem cells (ISCs) and progenitor cells of the fly midgut. RalA was required within ISCs for efficient regeneration downstream of Wnt signaling. Within the murine intestine, genetic deletion of either mammalian ortholog, Rala or Ralb, reduced ISC function and Lgr5 positivity, drove hypersensitivity to Wnt inhibition, and impaired tissue regeneration following damage. Ablation of both genes resulted in rapid crypt death. Mechanistically, RALA and RALB were required for efficient internalization of the Wnt receptor Frizzled-7. Together, we identify a conserved role for RAL GTPases in the promotion of optimal Wnt signaling, which defines ISC number and regenerative potential.
Collapse
Affiliation(s)
- Joel Johansson
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK
| | - Mate Naszai
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | | | | | - Bryan W Miller
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK
| | | | - Yachuan Yu
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | | | | | | | - Julia B Cordero
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK.
| | - Owen J Sansom
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK.
| |
Collapse
|
19
|
Deng C, Sun Y, Liu H, Wang W, Wang J, Zhang F. Selective adipogenic differentiation of human periodontal ligament stem cells stimulated with high doses of glucose. PLoS One 2018; 13:e0199603. [PMID: 29979705 PMCID: PMC6034828 DOI: 10.1371/journal.pone.0199603] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 06/11/2018] [Indexed: 02/07/2023] Open
Abstract
Periodontal tissue damage, accompanied by the degradation and destruction of periodontal tissue collagen, is one of the most clinically common complications and difficulty self-repair in patients with diabetes. Human periodontal ligament stem cells (PDLSC) are the undifferentiated mesenchymal cells that persist in the periodontal ligament after development of periodontal tissue and the ability of PDLSC osteogenic differentiation is responsible for repairing periodontal tissue defects. However, the reasons of high glucose environment in diabetic patients inhibiting PDLSC to repair periodontal tissues are unclear. To address these issues, we propose exposing PDLSC to high-sugar mimics the diabetic environment and investigating the activity of osteogenic differentiation and adipogenic differentiation of PDLSC. At the cellular level, high glucose can promote the adipogenic differentiation and inhibit osteogenic differentiation to decrease the self-repair ability of PDLSC in periodontal tissues. Mechanistically at the molecular level, these effects are elicited via regulating the mRNA and protein expression of C/EBPβ, PPAR-γ.
Collapse
Affiliation(s)
- Chao Deng
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University,Nanjing, China
- School of Stomatology, Wannan Medical College, Wuhu, China
| | - Yi Sun
- School of Stomatology, Wannan Medical College, Wuhu, China
| | - Hai Liu
- School of Stomatology, Wannan Medical College, Wuhu, China
| | - Wei Wang
- School of Stomatology, Wannan Medical College, Wuhu, China
| | - Jingmen Wang
- School of Stomatology, Wannan Medical College, Wuhu, China
| | - Feimin Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University,Nanjing, China
- * E-mail:
| |
Collapse
|
20
|
da Silveira Campos RM, Dâmaso AR, Masquio DCL, Duarte FO, Sene-Fiorese M, Aquino AE, Savioli FA, Quintiliano PCL, Kravchychyn ACP, Guimarães LI, Tock L, Oyama LM, Boldarine VT, Bagnato VS, Parizotto NA. The effects of exercise training associated with low-level laser therapy on biomarkers of adipose tissue transdifferentiation in obese women. Lasers Med Sci 2018; 33:1245-1254. [PMID: 29473115 DOI: 10.1007/s10103-018-2465-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 02/07/2018] [Indexed: 02/07/2023]
Abstract
Investigations suggest the benefits of low-level laser therapy (LLLT) to improve noninvasive body contouring treatments, inflammation, insulin resistance and to reduce body fat. However, the mechanism for such potential effects in association with exercise training (ET) and possible implications in browning adiposity processes remains unclear. Forty-nine obese women were involved, aged between 20 and 40 years with a body mass index (BMI) of 30-40 kg/m2. The volunteers were divided into Phototherapy (808 nm) and SHAM groups. Interventions consisted of exercise training and phototherapy applications post exercise for 4 months, with three sessions/week. Body composition, lipid profile, insulin resistance, atrial natriuretic peptide (ANP), WNT5 signaling, interleukin-6 (IL-6), and fibroblast growth factor-21 (FGF-21) were measured. Improvements in body mass, BMI, body fat mass, lean mass, visceral fat, waist circumference, insulin, HOMA-IR, total cholesterol, LDL-cholesterol, triglycerides, and ANP in both groups were demonstrated. Only the Phototherapy group showed a reduction in interleukin-6 and an increase in WNT5 signaling. In addition, it was possible to observe a higher magnitude change for the fat mass, insulin, HOMA-IR, and FGF-21 variables in the Phototherapy group. In the present investigation, it was demonstrated that exercise training associated with LLLT promotes an improvement in body composition and inflammatory processes as previously demonstrated. The Phototherapy group especially presented positive modifications of WNT5 signaling, FGF-21, and ANP, possible biomarkers associated with browning adiposity processes. This suggests that this kind of intervention promotes results applicable in clinical practice to control obesity and related comorbidities.
Collapse
Affiliation(s)
- Raquel Munhoz da Silveira Campos
- Department of Physiotherapy, Therapeutic Resources Laboratory, Universidade Federal de São Carlos (UFSCar), Rodovia Washington Luis, Km 235, São Carlos, São Paulo, 13565-905, Brazil.
| | - Ana Raimunda Dâmaso
- Post Graduated Program of Nutrition Paulista Medicine School, Universidade Federal de São Paulo (UNIFESP), Rua Marselhesa, 650-Vila Clementino, São Paulo, SP, 04020-050, Brazil.
| | | | - Fernanda Oliveira Duarte
- Electrical Engineering Department, Engineering School of São Carlos, Universidade de São Paulo (USP), Av. Trabalhador Sãocarlense 400, São Carlos, SP, 13566-590, Brazil
| | - Marcela Sene-Fiorese
- São Carlos Institute of Physics, Universidade de São Paulo (USP), PO Box 369, São Carlos, SP, 13560-970, Brazil
| | - Antonio Eduardo Aquino
- São Carlos Institute of Physics, Universidade de São Paulo (USP), PO Box 369, São Carlos, SP, 13560-970, Brazil
| | - Filippo Aragão Savioli
- Centro de Traumato-Ortopedia do Esporte (CETE), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | | | - Ana Claudia Pelissari Kravchychyn
- Post Graduated Program of Nutrition Paulista Medicine School, Universidade Federal de São Paulo (UNIFESP), Rua Marselhesa, 650-Vila Clementino, São Paulo, SP, 04020-050, Brazil
| | - Liliane Isabel Guimarães
- Centro de Traumato-Ortopedia do Esporte (CETE), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Lian Tock
- Weight Science, São Paulo, SP, Brazil
| | - Lila Missae Oyama
- Post Graduated Program of Nutrition Paulista Medicine School, Universidade Federal de São Paulo (UNIFESP), Rua Marselhesa, 650-Vila Clementino, São Paulo, SP, 04020-050, Brazil.,Department of Physiology Paulista Medicine School, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Valter Tadeu Boldarine
- Department of Physiology Paulista Medicine School, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Vanderlei Salvador Bagnato
- São Carlos Institute of Physics, Universidade de São Paulo (USP), PO Box 369, São Carlos, SP, 13560-970, Brazil.,Post Graduated Program of Biotechnology, Universidade Federal de São Carlos (UFSCar), São Carlos, SP, 13565-905, Brazil
| | - Nivaldo Antonio Parizotto
- Department of Physiotherapy, Therapeutic Resources Laboratory, Universidade Federal de São Carlos (UFSCar), Rodovia Washington Luis, Km 235, São Carlos, São Paulo, 13565-905, Brazil. .,Post Graduated Program of Biotechnology, Universidade Federal de São Carlos (UFSCar), São Carlos, SP, 13565-905, Brazil.
| |
Collapse
|
21
|
Abstract
Vitamin D, a secosteroid predominately obtained by endogenous production, has in recent years been linked to obesity and its comorbidities. The purpose of this review is to draw conclusions from animal and human studies on the effects of vitamin D on adipogenesis to identify the molecular links between vitamin D and obesity. The information presented herein was obtained from 4 databases (PubMed, CINAHL, Cochrane Library, Scopus) using predefined search terms, as well as research literature and other reviews. The effects of vitamin D on adipogenesis have been researched in several animal models, and the majority of these studies suggest vitamin D plays an inhibitory role in adipogenesis. Studies into vitamin D status and obesity in humans are limited, with the majority being observational epidemiological studies that provide no conclusions on cause and effect or clear links on the molecular mechanisms. The few cell culture and supplementation studies that have investigated adipogenesis in human cells indicate that, in contrast to findings from rodent studies, vitamin D is proadipogenic. There is insufficient evidence to determine whether 1) vitamin D deficiency is associated with a lean or obese phenotype, 2) vitamin D deficiency is a consequence of obesity, or (3) the effects of vitamin D on fat tissue are due to interactions with calcium.
Collapse
Affiliation(s)
- Clare F Dix
- Centre for Dietetic Research, School of Human Movement and Nutrition Sciences, University of Queensland, Brisbane, Australia
| | | | - Olivia R L Wright
- Centre for Dietetic Research, School of Human Movement and Nutrition Sciences, University of Queensland, Brisbane, Australia
| |
Collapse
|
22
|
Bai P, Xia N, Sun H, Kong Y. Pleiotrophin, a target of miR-384, promotes proliferation, metastasis and lipogenesis in HBV-related hepatocellular carcinoma. J Cell Mol Med 2017; 21:3023-3043. [PMID: 28557334 PMCID: PMC5661149 DOI: 10.1111/jcmm.13213] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 03/29/2017] [Indexed: 12/27/2022] Open
Abstract
Hepatitis B virus (HBV) infection plays a crucial role and is a major cause of hepatocellular carcinoma (HCC) in China. microRNAs (miRNAs) have emerged as key players in hepatic steatosis and carcinogenesis. We found that down-regulation of miR-384 expression was a common event in HCC, especially HBV-related HCC. However, the possible function of miR-384 in HBV-related HCC remains unclear. The oncogene pleiotrophin (PTN) was a target of miR-384. HBx inhibited miR-384, increasing PTN expression. The PTN receptor N-syndecan was highly expressed in HCC. PTN induced by HBx acted as a growth factor via N-syndecan on hepatocytes and further promoted cell proliferation, metastasis and lipogenesis. PTN up-regulated sterol regulatory element-binding protein 1c (SREBP-1c) through the N-syndecan/PI3K/Akt/mTORC1 pathway and the expression of lipogenic genes, including fatty acid synthesis (FAS). PTN-mediated de novo lipid synthesis played an important role in HCC proliferation and metastasis. PI3K/AKT and an mTORC1 inhibitor diminished PTN-induced proliferation, metastasis and lipogenesis. Taken together, these data strongly suggest that the dysregulation of miR-384 could play a crucial role in HBV related to HCC, and the target gene of miR-384, PTN, represents a new potential therapeutic target for the prevention of hepatic steatosis and further progression to HCC after chronic HBV infection.
Collapse
Affiliation(s)
- Pei‐song Bai
- Department of OncologyFirst Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Nan Xia
- Institute of Cancer Prevention and ControlPeking University Cancer HospitalBei'jingChina
| | - Hong Sun
- Department of OncologyFirst Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Ying Kong
- Department of OncologyFirst Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| |
Collapse
|
23
|
Martini CN, Gabrielli M, Bonifacino G, Codesido MM, Vila MDC. Lead enhancement of 3T3-L1 fibroblasts differentiation to adipocytes involves ERK, C/EBPβ and PPARγ activation. Mol Cell Biochem 2017. [DOI: 10.1007/s11010-017-3093-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
24
|
Reed KM, Mendoza KM, Abrahante JE, Barnes NE, Velleman SG, Strasburg GM. Response of turkey muscle satellite cells to thermal challenge. I. transcriptome effects in proliferating cells. BMC Genomics 2017; 18:352. [PMID: 28477619 PMCID: PMC5420122 DOI: 10.1186/s12864-017-3740-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 04/27/2017] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Climate change poses a multi-dimensional threat to food and agricultural systems as a result of increased risk to animal growth, development, health, and food product quality. This study was designed to characterize transcriptional changes induced in turkey muscle satellite cells cultured under cold or hot thermal challenge to better define molecular mechanisms by which thermal stress alters breast muscle ultrastructure. RESULTS Satellite cells isolated from the pectoralis major muscle of 7-weeks-old male turkeys from two breeding lines (16 weeks body weight-selected and it's randombred control) were proliferated in culture at 33 °C, 38 °C or 43 °C for 72 h. Total RNA was isolated and 12 libraries subjected to RNAseq analysis. Statistically significant differences in gene expression were observed among treatments and between turkey lines with a greater number of genes altered by cold treatment than by hot and fewer differences observed between lines than between temperatures. Pathway analysis found that cold treatment resulted in an overrepresentation of genes involved in cell signaling/signal transduction and cell communication/cell signaling as compared to control (38 °C). Heat-treated muscle satellite cells showed greater tendency towards expression of genes related to muscle system development and differentiation. CONCLUSIONS This study demonstrates significant transcriptome effects on turkey skeletal muscle satellite cells exposed to thermal challenge. Additional effects on gene expression could be attributed to genetic selection for 16 weeks body weight (muscle mass). New targets are identified for further research on the differential control of satellite cell proliferation in poultry.
Collapse
Affiliation(s)
- Kent M. Reed
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN USA
| | - Kristelle M. Mendoza
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN USA
| | - Juan E. Abrahante
- University of Minnesota Informatics Institute, University of Minnesota, Minneapolis, MN USA
| | - Natalie E. Barnes
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN USA
| | - Sandra G. Velleman
- Department of Animal Sciences, The Ohio State University, Columbus, OH USA
- Ohio Agricultural Research and Development Center, Wooster, OH USA
| | - Gale M. Strasburg
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI USA
| |
Collapse
|
25
|
Roh SY, Park JC. The role of nuclear factor I-C in tooth and bone development. J Korean Assoc Oral Maxillofac Surg 2017; 43:63-69. [PMID: 28462188 PMCID: PMC5410429 DOI: 10.5125/jkaoms.2017.43.2.63] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 04/06/2017] [Indexed: 01/15/2023] Open
Abstract
Nuclear factor I-C (NFI-C) plays a pivotal role in various cellular processes such as odontoblast and osteoblast differentiation. Nfic-deficient mice showed abnormal tooth and bone formation. The transplantation of Nfic-expressing mouse bone marrow stromal cells rescued the impaired bone formation in Nfic-/- mice. Studies suggest that NFI-C regulate osteogenesis and dentinogenesis in concert with several factors including transforming growth factor-β1, Krüppel-like factor 4, and β-catenin. This review will focus on the function of NFI-C during tooth and bone formation and on the relevant pathways that involve NFI-C.
Collapse
Affiliation(s)
- Song Yi Roh
- Department of Oral Histology-Developmental Biology and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Korea
| | - Joo-Cheol Park
- Department of Oral Histology-Developmental Biology and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Korea
| |
Collapse
|
26
|
Lee J, Imm JY, Lee SH. β-Catenin Mediates Anti-adipogenic and Anticancer Effects of Arctigenin in Preadipocytes and Breast Cancer Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:2513-2520. [PMID: 28279068 DOI: 10.1021/acs.jafc.7b00112] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Arctigenin is a lignan abundant in Asteraceae plants and has anti-inflammatory, antiobesity, and anticancer activities. Obesity is one of the leading causes of several types of cancers including breast cancer. The current study was performed to investigate if arctigenin suppresses differentiation of preadipocytes and proliferation of breast cancer cells and to explore potential molecular mechanisms. Treatment of arctigenin reduced lipid accumulation in differentiated 3T3-L1 adipocytes in a dose- and time-dependent manner without toxicity. Arctigenin suppressed the expression of peroxisome proliferator-activated receptor-gamma (PPARγ), CCAAT/enhancer-binding protein-alpha (C/EBPα), perilipin, and fatty acid binding protein 4 (FABP4) in a dose-dependent manner in differentiated 3T3-L1 cells. Both total and unphosphorylated (active) β-catenin were increased in whole cell lysates and the nuclear fraction of differentiated 3T3-L1 cells treated with 25 μM arctigenin. On the other hand, arctigenin decreased proliferation of two human breast cancer cells (MCF-7 and MDA-MB-231). Arctigenin induced apoptosis and decreased expression of total and unphosphorylated (active) β-catenin and cyclin D1 in MCF-7, but not in MDA-MB-231. These data indicate that arctigenin suppressed adipogenesis in preadipocytes and activated apoptosis in estrogen receptor (ER) positive breast cancer cells through modulating expression of β-catenin.
Collapse
Affiliation(s)
- Jihye Lee
- Department of Nutrition and Food Science, University of Maryland , College Park, Maryland 20742, United States
| | - Jee-Young Imm
- Department of Foods and Nutrition, Kookmin University , Seoul 136-702, Korea
| | - Seong-Ho Lee
- Department of Nutrition and Food Science, University of Maryland , College Park, Maryland 20742, United States
| |
Collapse
|
27
|
Mota de Sá P, Richard AJ, Hang H, Stephens JM. Transcriptional Regulation of Adipogenesis. Compr Physiol 2017; 7:635-674. [PMID: 28333384 DOI: 10.1002/cphy.c160022] [Citation(s) in RCA: 256] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Adipocytes are the defining cell type of adipose tissue. Once considered a passive participant in energy storage, adipose tissue is now recognized as a dynamic organ that contributes to several important physiological processes, such as lipid metabolism, systemic energy homeostasis, and whole-body insulin sensitivity. Therefore, understanding the mechanisms involved in its development and function is of great importance. Adipocyte differentiation is a highly orchestrated process which can vary between different fat depots as well as between the sexes. While hormones, miRNAs, cytoskeletal proteins, and many other effectors can modulate adipocyte development, the best understood regulators of adipogenesis are the transcription factors that inhibit or promote this process. Ectopic expression and knockdown approaches in cultured cells have been widely used to understand the contribution of transcription factors to adipocyte development, providing a basis for more sophisticated in vivo strategies to examine adipogenesis. To date, over two dozen transcription factors have been shown to play important roles in adipocyte development. These transcription factors belong to several families with many different DNA-binding domains. While peroxisome proliferator-activated receptor gamma (PPARγ) is undoubtedly the most important transcriptional modulator of adipocyte development in all types of adipose tissue, members of the CCAAT/enhancer-binding protein, Krüppel-like transcription factor, signal transducer and activator of transcription, GATA, early B cell factor, and interferon-regulatory factor families also regulate adipogenesis. The importance of PPARγ activity is underscored by several covalent modifications that modulate its activity and its ability to modulate adipocyte development. This review will primarily focus on the transcriptional control of adipogenesis in white fat cells and on the mechanisms involved in this fine-tuned developmental process. © 2017 American Physiological Society. Compr Physiol 7:635-674, 2017.
Collapse
Affiliation(s)
- Paula Mota de Sá
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Allison J Richard
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Hardy Hang
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Jacqueline M Stephens
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| |
Collapse
|
28
|
Transcriptional regulation analysis of FAM3A gene and its effect on adipocyte differentiation. Gene 2016; 595:92-98. [DOI: 10.1016/j.gene.2016.09.038] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 09/04/2016] [Accepted: 09/24/2016] [Indexed: 11/17/2022]
|
29
|
Jeon M, Rahman N, Kim YS. Wnt/β-catenin signaling plays a distinct role in methyl gallate-mediated inhibition of adipogenesis. Biochem Biophys Res Commun 2016; 479:22-7. [PMID: 27592552 DOI: 10.1016/j.bbrc.2016.08.178] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 08/31/2016] [Indexed: 01/14/2023]
Abstract
The canonical Wnt/β-catenin signaling not only features in many developmental processes but also recently emerged as an attractive negative regulator of differentiation of preadipocytes into adipocytes. Here, we show that β-catenin signaling plays a distinct role in methyl gallate (MG)-mediated inhibition of 3T3-L1 adipocytes differentiation. We found that the expression of β-catenin decreased after adipogenic hormonal induction, whereas incubation of the differentiating cells with a physiological concentration of MG during adipogenic hormonal induction significantly prevented β-catenin degradation by activating Wnt signaling components such as Wnt1, Wnt10b, Fzd1, Fzd2, Lrp5, Lrp6, Dvl1, and Dvl2. Mechanistic experiments revealed that MG treatment during early adipocytic differentiation specifically inhibited degradation of β-catenin caused by phosphorylation at serine-33. In addition, MG treatment led to phosphorylation of GSK3β, which is one of β-catenin-degrading enzymes. Consequently, MG treatment facilitated translocation of the stabilized β-catenin from the cytoplasm to nucleus, and activates its target genes cyclin D1 and c-Myc. Furthermore, MG-induced stabilization of β-catenin suppresses PPARγ expression. Moreover, pharmacological activation or inhibition of β-catenin signaling during adipocytes differentiation decreased and increased, respectively, the level of the key adipogenic marker, PPARγ, and of its downstream targets, aP2 and adiponectin while MG treatment effectively reversed their expression level. Collectively, our data suggest that MG is a novel pharmacological stimulator of canonical Wnt/β-catenin signaling, and therefore represents a promising therapeutic agent in obesity.
Collapse
Affiliation(s)
- Miso Jeon
- Department of Microbiology, College of Medicine, Soonchunhyang University, South Korea; Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, South Korea
| | - Naimur Rahman
- Department of Microbiology, College of Medicine, Soonchunhyang University, South Korea; Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, South Korea
| | - Yong-Sik Kim
- Department of Microbiology, College of Medicine, Soonchunhyang University, South Korea; Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, South Korea.
| |
Collapse
|
30
|
Brioche T, Pagano AF, Py G, Chopard A. Muscle wasting and aging: Experimental models, fatty infiltrations, and prevention. Mol Aspects Med 2016; 50:56-87. [PMID: 27106402 DOI: 10.1016/j.mam.2016.04.006] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 04/13/2016] [Accepted: 04/13/2016] [Indexed: 12/21/2022]
Abstract
Identification of cost-effective interventions to maintain muscle mass, muscle strength, and physical performance during muscle wasting and aging is an important public health challenge. It requires understanding of the cellular and molecular mechanisms involved. Muscle-deconditioning processes have been deciphered by means of several experimental models, bringing together the opportunities to devise comprehensive analysis of muscle wasting. Studies have increasingly recognized the importance of fatty infiltrations or intermuscular adipose tissue for the age-mediated loss of skeletal-muscle function and emphasized that this new important factor is closely linked to inactivity. The present review aims to address three main points. We first mainly focus on available experimental models involving cell, animal, or human experiments on muscle wasting. We next point out the role of intermuscular adipose tissue in muscle wasting and aging and try to highlight new findings concerning aging and muscle-resident mesenchymal stem cells called fibro/adipogenic progenitors by linking some cellular players implicated in both FAP fate modulation and advancing age. In the last part, we review the main data on the efficiency and molecular and cellular mechanisms by which exercise, replacement hormone therapies, and β-hydroxy-β-methylbutyrate prevent muscle wasting and sarcopenia. Finally, we will discuss a potential therapeutic target of sarcopenia: glucose 6-phosphate dehydrogenase.
Collapse
Affiliation(s)
- Thomas Brioche
- Université de Montpellier, INRA, UMR 866 Dynamique Musculaire et Métabolisme, Montpellier F-34060, France.
| | - Allan F Pagano
- Université de Montpellier, INRA, UMR 866 Dynamique Musculaire et Métabolisme, Montpellier F-34060, France
| | - Guillaume Py
- Université de Montpellier, INRA, UMR 866 Dynamique Musculaire et Métabolisme, Montpellier F-34060, France
| | - Angèle Chopard
- Université de Montpellier, INRA, UMR 866 Dynamique Musculaire et Métabolisme, Montpellier F-34060, France
| |
Collapse
|
31
|
Functional transcription factor target discovery via compendia of binding and expression profiles. Sci Rep 2016; 6:20649. [PMID: 26857150 PMCID: PMC4746627 DOI: 10.1038/srep20649] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 01/07/2016] [Indexed: 12/21/2022] Open
Abstract
Genome-wide experiments to map the DNA-binding locations of transcription-associated factors (TFs) have shown that the number of genes bound by a TF far exceeds the number of possible direct target genes. Distinguishing functional from non-functional binding is therefore a major challenge in the study of transcriptional regulation. We hypothesized that functional targets can be discovered by correlating binding and expression profiles across multiple experimental conditions. To test this hypothesis, we obtained ChIP-seq and RNA-seq data from matching cell types from the human ENCODE resource, considered promoter-proximal and distal cumulative regulatory models to map binding sites to genes, and used a combination of linear and non-linear measures to correlate binding and expression data. We found that a high degree of correlation between a gene’s TF-binding and expression profiles was significantly more predictive of the gene being differentially expressed upon knockdown of that TF, compared to using binding sites in the cell type of interest only. Remarkably, TF targets predicted from correlation across a compendium of cell types were also predictive of functional targets in other cell types. Finally, correlation across a time course of ChIP-seq and RNA-seq experiments was also predictive of functional TF targets in that tissue.
Collapse
|
32
|
Poulos SP, Dodson MV, Culver MF, Hausman GJ. The increasingly complex regulation of adipocyte differentiation. Exp Biol Med (Maywood) 2015; 241:449-56. [PMID: 26645953 DOI: 10.1177/1535370215619041] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 10/29/2015] [Indexed: 01/31/2023] Open
Abstract
Adipose (AD) tissue development and function relies on the ability of adipocytes to proliferate and differentiate into lipid-containing cells that also have endocrine function. Research suggests that certain conditions can induce AD tissue stem cells to differentiate into various cell types and that the microenvironment of the cell, including the extracellular matrix (ECM), is essential in maintaining cell and tissue function. This review provides an overview of factors involved in the proliferation and differentiation of adipocytes. A brief review of the numerous factors that influence PPARγ, the transcription factor thought to be the master regulator of adipocyte differentiation, provides context of established pathways that regulate adipogenesis. Thought provoking findings from research with hypoxia that is supported by earlier research that vascular development is related to adipogenesis are reviewed. Finally, our understanding of the critical role of the ECM and environment in adipogenesis is discussed and compared with studies that suggest that adipocytes may dedifferentiate and can convert into other cell types.
Collapse
Affiliation(s)
| | - Michael V Dodson
- Department of Animal Science, Washington State University, Pullman, WA 99164, USA
| | | | - Gary J Hausman
- Animal and Dairy Science Department, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
33
|
Liu J, Yang P, Shi H, Sun X, Lee SH, Yu L(L. A novel Gynostemma pentaphyllum saponin and its adipogenesis inhibitory effect through modulating Wnt/β-catenin pathway and cell cycle in mitotic clonal expansion. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.06.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
|
34
|
Zhuang B, Luo X, Rao H, Li Q, Shan N, Liu X, Qi H. Oxidative stress-induced C/EBPβ inhibits β-catenin signaling molecule involving in the pathology of preeclampsia. Placenta 2015; 36:839-46. [PMID: 26166436 DOI: 10.1016/j.placenta.2015.06.016] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 06/16/2015] [Accepted: 06/30/2015] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Oxidative stress-induced trophoblast cell dysfunction is a major pathology in preeclampsia (PE). Recently, CCAAT/enhancer binding protein beta (C/EBPβ) has been investigated as a tumor suppressor that participates in tumor invasion. However, the function of C/EBPβ in trophoblast cells remains unknown. Our study was designed to detect the expression of C/EBPβ in the preeclamptic placenta and to identify the underlying mechanisms of oxidative stress. METHODS Human placental tissues with PE were collected. The expression of C/EBPβ and β-catenin were detected. Human first trimester extravillous trophoblast cell (HTR8/SVneo) line exposed to hypoxia/reoxygenation (H/R) was employed as an oxidative stress model in vitro to investigate the effects of C/EBPβ on invasion and the expression of β-catenin. Moreover, first trimester-derived placental villous explants were used to verify the effects of C/EBPβ and β-catenin in placentation. RESULTS In preeclamptic placentas, C/EBPβ was overexpressed and β-catenin was decreased. In addition, C/EBPβ was found to have increased expression in H/R-treated HTR8/SVneo cells and villous explants. C/EBPβ knockdown and β-catenin activation could significantly promote the invasion of HTR8/SVneo cells, enhance the outgrowth and migration in villous explants and inhibit the excessive generation of intracellular ROS. These findings might be related to the increased activities of MMP-2/9 and the decreased expression of TIMP-1/2. Meanwhile, C/EBPβ knockdown remarkably increased the expression of β-catenin. DISCUSSION We hypothesize that the oxidative stress-induced overexpression of C/EBPβ might influence the activity of MMPs by regulating the Wnt/β-catenin signaling pathway to affect the invasion of trophoblast cells, which then participate in the pathogenesis of preeclampsia.
Collapse
Affiliation(s)
- B Zhuang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Yixue Road, Yuzhong District, Chongqing 400016, China
| | - X Luo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Yixue Road, Yuzhong District, Chongqing 400016, China
| | - H Rao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Yixue Road, Yuzhong District, Chongqing 400016, China
| | - Q Li
- Department of Pathology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Yixue Road, Yuzhong District, Chongqing 400016, China
| | - N Shan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Yixue Road, Yuzhong District, Chongqing 400016, China
| | - X Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Yixue Road, Yuzhong District, Chongqing 400016, China
| | - H Qi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Yixue Road, Yuzhong District, Chongqing 400016, China.
| |
Collapse
|
35
|
Hu L, Yang G, Hägg D, Sun G, Ahn JM, Jiang N, Ricupero CL, Wu J, Rodhe CH, Ascherman JA, Chen L, Mao JJ. IGF1 Promotes Adipogenesis by a Lineage Bias of Endogenous Adipose Stem/Progenitor Cells. Stem Cells 2015; 33:2483-95. [PMID: 26010009 DOI: 10.1002/stem.2052] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 10/31/2014] [Indexed: 01/08/2023]
Abstract
Adipogenesis is essential for soft tissue reconstruction following trauma or tumor resection. We demonstrate that CD31(-)/34(+)/146(-) cells, a subpopulation of the stromal vascular fraction (SVF) of human adipose tissue, were robustly adipogenic. Insulin growth factor-1 (IGF1) promoted a lineage bias towards CD31(-)/34(+)/146(-) cells at the expense of CD31(-)/34(+)/146(+) cells. IGF1 was microencapsulated in poly(lactic-co-glycolic acid) scaffolds and implanted in the inguinal fat pad of C57Bl6 mice. Control-released IGF1 induced remarkable adipogenesis in vivo by recruiting endogenous cells. In comparison with the CD31(-)/34(+)/146(+) cells, CD31(-)/34(+)/146(-) cells had a weaker Wnt/β-catenin signal. IGF1 attenuated Wnt/β-catenin signaling by activating Axin2/PPARγ pathways in SVF cells, suggesting IGF1 promotes CD31(-)/34(+)/146(-) bias through tuning Wnt signal. PPARγ response element (PPRE) in Axin2 promoter was crucial for Axin2 upregulation, suggesting that PPARγ transcriptionally activates Axin2. Together, these findings illustrate an Axin2/PPARγ axis in adipogenesis that is particularly attributable to a lineage bias towards CD31(-)/34(+)/146(-) cells, with implications in adipose regeneration.
Collapse
Affiliation(s)
- Li Hu
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China.,Center for Craniofacial Regeneration (CCR), New York, New York, USA
| | - Guodong Yang
- Center for Craniofacial Regeneration (CCR), New York, New York, USA
| | - Daniel Hägg
- Center for Craniofacial Regeneration (CCR), New York, New York, USA
| | - Guoming Sun
- Center for Craniofacial Regeneration (CCR), New York, New York, USA
| | - Jeffrey M Ahn
- Department of Otolaryngology, New York, New York, USA
| | - Nan Jiang
- Center for Craniofacial Regeneration (CCR), New York, New York, USA
| | | | - June Wu
- Department of Plastic Surgery, Columbia University Medical Center, New York, New York, USA
| | - Christine Hsu Rodhe
- Department of Plastic Surgery, Columbia University Medical Center, New York, New York, USA
| | - Jeffrey A Ascherman
- Department of Plastic Surgery, Columbia University Medical Center, New York, New York, USA
| | - Lili Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Jeremy J Mao
- Center for Craniofacial Regeneration (CCR), New York, New York, USA
| |
Collapse
|
36
|
CCAAT/enhancer binding protein β in relation to ER stress, inflammation, and metabolic disturbances. BIOMED RESEARCH INTERNATIONAL 2015; 2015:324815. [PMID: 25699273 PMCID: PMC4324884 DOI: 10.1155/2015/324815] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 12/12/2014] [Accepted: 12/12/2014] [Indexed: 12/12/2022]
Abstract
The prevalence of the metabolic syndrome and underlying metabolic disturbances increase rapidly in developed countries. Various molecular targets are currently under investigation to unravel the molecular mechanisms that cause these disturbances. This is done in attempt to counter or prevent the negative health consequences of the metabolic disturbances. Here, we reviewed the current knowledge on the role of C/EBP-β in these metabolic disturbances. C/EBP-β deletion in mice resulted in downregulation of hepatic lipogenic genes and increased expression of β-oxidation genes in brown adipose tissue. Furthermore, C/EBP-β is important in the differentiation and maturation of adipocytes and is increased during ER stress and proinflammatory conditions. So far, studies were only conducted in animals and in cell systems. The results found that C/EBP-β is an important transcription factor within the metabolic disturbances of the metabolic system. Therefore, it is interesting to examine the potential role of C/EBP-β at molecular and physiological level in humans.
Collapse
|
37
|
In silico identification of conserved microRNAs and their targets in bovine fat tissue. Gene 2015; 559:119-28. [PMID: 25626178 DOI: 10.1016/j.gene.2015.01.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 01/09/2015] [Accepted: 01/11/2015] [Indexed: 12/22/2022]
Abstract
MicroRNAs (miRNAs) represent a newly identified class of non-protein-coding ~22 nt small RNA which plays important roles in multiple biological processes by degrading targeted mRNA or repressing mRNA translation. Here we present EST (expressed sequence tags)-based and GSS (Genomic Survey Sequences)-based combined approach for the detection of conserved miRNAs of cattle. A total of 20 conserved miRNAs that belong to 18 miRNA families were detected following a range of filtering criteria; their functions were further predicted and analyzed. To confirm our prediction, a miRNA-detecting microarray was designed with probes complementary to previously known mature miRNA sequences from 131 organisms. After hybridizing with small RNAs extracted from beef cattle subcutaneous fat tissue, 219 (32.30%) miRNAs were detected in the 679 known Bos taurus miRNAs and all the miRNAs predicted above were also detected. Conformation of 22 most abundant miRNA expression by qRT-PCR indicated that they were highly accumulated not only in subcutaneous fat tissue but also in intramuscular fat tissue. Bioinformatics of KEGG pathway analysis suggested that 4 differential expression miRNAs (miR-143, miR-145, miR-2325c and miR-2361) involved in different pathways and target genes may regulate the fat deposition differently. Taken together, our results expand the number of known bovine miRNAs and provide a thorough account of the miRNA transcriptome in bovine adipose tissue.
Collapse
|
38
|
Guo L, Li X, Tang QQ. Transcriptional regulation of adipocyte differentiation: a central role for CCAAT/enhancer-binding protein (C/EBP) β. J Biol Chem 2014; 290:755-61. [PMID: 25451943 DOI: 10.1074/jbc.r114.619957] [Citation(s) in RCA: 267] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
A detailed understanding of the processes controlling adipogenesis is instrumental in the fight against the obesity epidemic. Adipogenesis is controlled by a transcriptional cascade composed of a large number of transcriptional factors, among which CCAAT/enhancer-binding protein (C/EBP) β plays an essential role. During 3T3-L1 adipocyte differentiation, C/EBPβ is induced early to transactivate the expression of C/EBPα and peroxisome proliferator-activated receptor γ (PPARγ), two master transcription factors for terminal adipocyte differentiation. Studies in recent years have revealed many new target genes of C/EBPβ, implicating its participation in many other processes during adipogenesis, such as mitotic clonal expansion, epigenetic regulation, unfolded protein response, and autophagy. Moreover, the function of C/EBPβ is highly regulated by post-translational modifications, which are crucial for the proper activation of the adipogenic program. Advances toward elucidation of the function and roles of the post-translational modification of C/EBPβ during adipogenesis will greatly improve our understanding of the molecular mechanisms governing adipogenesis.
Collapse
Affiliation(s)
- Liang Guo
- From the Key Laboratory of Metabolism and Molecular Medicine, the Ministry of Education and Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Xi Li
- From the Key Laboratory of Metabolism and Molecular Medicine, the Ministry of Education and Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Qi-Qun Tang
- From the Key Laboratory of Metabolism and Molecular Medicine, the Ministry of Education and Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai 200032, China
| |
Collapse
|
39
|
Strakovsky RS, Lezmi S, Flaws JA, Schantz SL, Pan YX, Helferich WG. Genistein exposure during the early postnatal period favors the development of obesity in female, but not male rats. Toxicol Sci 2013; 138:161-74. [PMID: 24361872 DOI: 10.1093/toxsci/kft331] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Genistein (Gen), the primary isoflavone in soy, has been shown to adversely affect various endocrine-mediated endpoints in rodents and humans. Soy formula intake by human infants has been associated with early age at menarche and decreased female-typical behavior in girls. Adipose deposition and expansion are also hormonally regulated and Gen has been shown to alter these processes. However, little is known about the impact of early-life soy intake on metabolic homeostasis in adulthood. The current study examined the impact of early-life Gen exposure on adulthood body composition (by magnetic resonance imaging) and the molecular signals mediating adipose expansion. From postnatal day (PND) 1 to 22, rat pups were daily orally dosed with 50mg/kg Gen to mimic blood Gen levels in human infants fed soy formula. Female but not male Gen-exposed rats had increased fat/lean mass ratio, fat mass, adipocyte size and number, and decreased muscle fiber perimeter. PND22 Gen-exposed females, but not males, had increased expression of adipogenic factors, including CCAAT/enhancer binding protein alpha (Cebpα), CCAAT/enhancer binding protein beta (Cebpβ), and peroxisome proliferator-activated receptor gamma (Pparγ). Furthermore, Wingless-related MMTV integration site 10b (Wnt10b), a critical regulator of adipogenic cell fate determination, was hypermethylated and had decreased expression in adipose of PND22 Gen-exposed females. These data suggest that developmental Gen exposure in rats has gender-specific effects on adiposity that closely parallel the effects of a postweaning high-fat diet and underscore the importance of considering timing of exposure and gender when establishing safety recommendations for early-life dietary Gen intake.
Collapse
|
40
|
F-box only protein 9 is required for adipocyte differentiation. Biochem Biophys Res Commun 2013; 435:239-43. [PMID: 23643813 DOI: 10.1016/j.bbrc.2013.04.072] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 04/26/2013] [Indexed: 11/24/2022]
Abstract
The objective of this study is to investigate whether F-box only protein 9 (FBXO9), an ubiquitination E3 ligase, has a functional role in adipocyte differentiation. Expression of FBXO9 was compared between obese mice and control lean mice using real-time PCR. Also, expression pattern of FBXO9 was monitored during 3T3-L1 adipocyte differentiation. FBXO9 was highly expressed in obese mice, and increased in the early stages of adipogenesis. To verify a functional role of FBXO9 in adipogenesis, FBXO9 was knocked down using transfection of siRNAs against FBXO9 into 3T3-L1 cells during the induction of adipogenesis. Knockdown of FBXO9 in early stage of adipogenesis almost completely inhibited adipogenesis, and CCAAT/enhancer binding protein β (C/EBPβ) levels were significantly reduced. However, the cells stably expressing C/EBPβ were fairly differentiated into adipocytes in the FBXO9 knockdown condition. These results suggest that FBXO9 is required for adipocyte differentiation, and C/EBPβ plays a role in the effect of FBXO9 on adipogenesis.
Collapse
|
41
|
Cho YM, Kim DH, Kwak SN, Jeong SW, Kwon OJ. X-box binding protein 1 enhances adipogenic differentiation of 3T3-L1 cells through the downregulation of Wnt10b expression. FEBS Lett 2013; 587:1644-9. [PMID: 23603388 DOI: 10.1016/j.febslet.2013.04.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 04/01/2013] [Accepted: 04/08/2013] [Indexed: 11/24/2022]
Abstract
Differentiation of preadipocytes into adipocytes is controlled by various transcription factors. Recently, the pro-adipogenic function of XBP1, a transcription factor upregulated by endoplasmic reticulum stress, has been reported. In this study, we demonstrated that XBP1 suppresses the expression of Wnt10b, an anti-adipogenic Wnt, during the differentiation of 3T3-L1 preadipocytes. The expression pattern of XBP1 was reciprocal to that of Wnt10b during the early stage of adipogenesis. The intracellular protein levels of β-catenin were negatively regulated by XBP1. Direct binding of XBP1 to the Wnt10b promoter and the subsequent decrease of the β-catenin signalling pathway represent a novel adipogenic differentiation mechanism.
Collapse
Affiliation(s)
- Yoon Mi Cho
- Department of Biochemistry, The Catholic University of Korea, Seoul 137-701, Republic of Korea
| | | | | | | | | |
Collapse
|
42
|
Role of C/EBPβ-LAP and C/EBPβ-LIP in early adipogenic differentiation of human white adipose-derived progenitors and at later stages in immature adipocytes. Differentiation 2013; 85:20-31. [DOI: 10.1016/j.diff.2012.11.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Revised: 09/20/2012] [Accepted: 11/26/2012] [Indexed: 12/20/2022]
|
43
|
Unique responses of stem cell-derived vascular endothelial and mesenchymal cells to high levels of glucose. PLoS One 2012; 7:e38752. [PMID: 22701703 PMCID: PMC3368917 DOI: 10.1371/journal.pone.0038752] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 05/11/2012] [Indexed: 12/21/2022] Open
Abstract
Diabetes leads to complications in selected organ systems, and vascular endothelial cell (EC) dysfunction and loss is the key initiating and perpetuating step in the development of these complications. Experimental and clinical studies have shown that hyperglycemia leads to EC dysfunction in diabetes. Vascular stem cells that give rise to endothelial progenitor cells (EPCs) and mesenchymal progenitor cells (MPCs) represent an attractive target for cell therapy for diabetic patients. Whether these vascular stem/progenitor cells succumb to the adverse effects of high glucose remains unknown. We sought to determine whether adult vascular stem/progenitor cells display cellular activation and dysfunction upon exposure to high levels of glucose as seen in diabetic complications. Mononuclear cell fraction was prepared from adult blood and bone marrow. EPCs and MPCs were derived, characterized, and exposed to either normal glucose (5 mmol/L) or high glucose levels (25 mmol/L). We then assayed for cell activity and molecular changes following both acute and chronic exposure to high glucose. Our results show that high levels of glucose do not alter the derivation of either EPCs or MPCs. The adult blood-derived EPCs were also resistant to the effects of glucose in terms of growth. Acute exposure to high glucose levels increased caspase-3 activity in EPCs (1.4x increase) and mature ECs (2.3x increase). Interestingly, MPCs showed a transient reduction in growth upon glucose challenge. Our results also show that glucose skews the differentiation of MPCs towards the adipocyte lineage while suppressing other mesenchymal lineages. In summary, our studies show that EPCs are resistant to the effects of high levels of glucose, even following chronic exposure. The findings further show that hyperglycemia may have detrimental effects on the MPCs, causing reduced growth and altering the differentiation potential.
Collapse
|