1
|
Jang B, Kim Y, Song J, Kim YW, Lee WY. Identifying Herbal Candidates and Active Ingredients Against Postmenopausal Osteoporosis Using Biased Random Walk on a Multiscale Network. Int J Mol Sci 2024; 25:12322. [PMID: 39596387 PMCID: PMC11594441 DOI: 10.3390/ijms252212322] [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: 11/01/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Postmenopausal osteoporosis is a major global health concern, particularly affecting aging women, and necessitates innovative treatment options. Herbal medicine, with its multi-compound, multi-target characteristics, offers a promising approach for complex diseases. In this study, we applied multiscale network and random walk-based analyses to identify candidate herbs and their active ingredients for postmenopausal osteoporosis, focusing on their underlying mechanisms. A dataset of medicinal herbs, their active ingredients, and protein targets was compiled, and diffusion profiles were calculated to assess the propagation effects. Through correlation analysis, we prioritized herbs based on their relevance to osteoporosis, identifying the top candidates like Benincasae Semen, Glehniae Radix, Corydalis Tuber, and Houttuyniae Herba. Gene Set Enrichment Analysis (GSEA) revealed that the 49 core protein targets of these herbs were significantly associated with pathways related to inflammation, osteoclast differentiation, and estrogen metabolism. Notably, compounds such as falcarindiol from Glehniae Radix and tetrahydrocoptisine from Corydalis Tuber-previously unstudied for osteoporosis-were predicted to interact with inflammation-related proteins, including IL6, IL1B, and TNF, affecting key biological processes like apoptosis and cell proliferation. This study advances the understanding of herbal therapies for osteoporosis and offers a framework for discovering novel therapeutic agents.
Collapse
Affiliation(s)
- Boyun Jang
- IntegroMediLab Co., Ltd., Seoul 04626, Republic of Korea
| | - Youngsoo Kim
- IntegroMediLab Co., Ltd., Seoul 04626, Republic of Korea
| | - Jungbin Song
- Department of Herbal Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Young-Woo Kim
- School of Korean Medicine, Dongguk University, Gyeongju 38066, Republic of Korea
| | - Won-Yung Lee
- School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea
- Research Center of Traditional Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea
| |
Collapse
|
2
|
Huang L, He S, Wang T, Long K, Ma B, Wu P, Gong Y, Zhong D, Yang Q, Wu J, Li X. circNDUFA13 stimulates adipogenesis of bone marrow-derived mesenchymal stem cells via interaction with STAT3. Sci Rep 2024; 14:19787. [PMID: 39187566 PMCID: PMC11347618 DOI: 10.1038/s41598-024-70867-9] [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: 01/30/2024] [Accepted: 08/22/2024] [Indexed: 08/28/2024] Open
Abstract
Circular RNAs (circRNAs) in controlling gene expression have been highlighted by increasing evidence, and their dysregulation has been linked to various diseases. However, the limited role of circRNAs in the adipogenesis of bone marrow-derived mesenchymal stem cells (BMSCs) has been explored. High-throughput sequencing of circRNA was carried out on BMSCs and AD induction 7d BMSCs. Then a substantial upregulation of circNDUFA13 was detected among circRNAs in AD induction 7d BMSCs. We found that the adipogenic differentiation of BMSCs was positively linked with circNDUFA13 expression levels. Adipogenesis in BMSCs was effectively inhibited by circNDUFA13 knockdown, whereas overexpression of circNDUFA13 promoted adipogenesis. It was noted that circNDUFA13 regulated the adipogenic differentiation of BMSCs by directly interacting with the signal transducer and activator of transcription 3 (STAT3), which activates CEBPβ transcription. The in vitro model also validated the in vivo findings. our results suggest that circNDUFA13 controlled the adipogenic differentiation of BMSCs by targeting STAT3 and CEBPβ activation.
Collapse
Affiliation(s)
- Longsheng Huang
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, 332005, Jiangxi, China
| | - Shan He
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, 332005, Jiangxi, China
| | - Tao Wang
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, 332005, Jiangxi, China
| | - Kai Long
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, 332005, Jiangxi, China
| | - Baicheng Ma
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, 332005, Jiangxi, China
| | - Ping Wu
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, 332005, Jiangxi, China
| | - Ying Gong
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, 332005, Jiangxi, China
| | - Donghuo Zhong
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, 332005, Jiangxi, China
| | - Qianyong Yang
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, 332005, Jiangxi, China
- Jiujiang Key Laboratory of Rare Disease ResearchSchool of Basic Medical Sciences, Jiujiang University, Jiujiang, 332005, China
| | - Jianfang Wu
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, 332005, Jiangxi, China
| | - Xingnuan Li
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, 332005, Jiangxi, China.
| |
Collapse
|
3
|
Abstract
Although osteoblasts and osteocytes are descended from the same lineage, they each have unique and essential roles in bone. Targeting gene deletion to osteoblasts and osteocytes using the Cre/loxP system has greatly increased our current understanding of how these cells function. Additionally, the use of the Cre/loxP system in conjunction with cell-specific reporters has enabled lineage tracing of these bone cells both in vivo and ex vivo. However, concerns have been raised regarding the specificity of the promoters used and the resulting off-target effects on cells within and outside of the bone. In this review, we have summarized the main mouse models that have been used to determine the functions of specific genes in osteoblasts and osteocytes. We discuss the expression patterns and specificity of the different promoter fragments during osteoblast to osteocyte differentiation in vivo. We also highlight how their expression in non-skeletal tissues may complicate the interpretation of study results. A thorough understanding of when and where these promoters are activated will enable improved study design and greater confidence in data interpretation.
Collapse
Affiliation(s)
- Y Kitase
- Indiana Center for Musculoskeletal Health, Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, United States of America
| | - M Prideaux
- Indiana Center for Musculoskeletal Health, Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, United States of America.
| |
Collapse
|
4
|
Li L, Wang Y, Wang Z, Xue D, Dai C, Gao X, Ma J, Hang K, Pan Z. Knockdown of FOXA1 enhances the osteogenic differentiation of human bone marrow mesenchymal stem cells partly via activation of the ERK1/2 signalling pathway. Stem Cell Res Ther 2022; 13:456. [PMID: 36064451 PMCID: PMC9446550 DOI: 10.1186/s13287-022-03133-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/11/2022] [Indexed: 11/21/2022] Open
Abstract
Background The available therapeutic options for large bone defects remain extremely limited, requiring new strategies to accelerate bone healing. Genetically modified bone mesenchymal stem cells (BMSCs) with enhanced osteogenic capacity are recognised as one of the most promising treatments for bone defects. Methods We performed differential expression analysis of miRNAs between human BMSCs (hBMSCs) and human dental pulp stem cells (hDPSCs) to identify osteogenic differentiation-related microRNAs (miRNAs). Furthermore, we identified shared osteogenic differentiation-related miRNAs and constructed an miRNA-transcription network. The Forkhead box protein A1 (FOXA1) knockdown strategy with a lentiviral vector was used to explore the role of FOXA1 in the osteogenic differentiation of MSCs. Cell Counting Kit-8 was used to determine the effect of the knockdown of FOXA1 on hBMSC proliferation; real-time quantitative reverse transcription PCR (qRT-PCR) and western blotting were used to investigate target genes and proteins; and alkaline phosphatase (ALP) staining and Alizarin Red staining (ARS) were used to assess ALP activity and mineral deposition, respectively. Finally, a mouse model of femoral defects was established in vivo, and histological evaluation and radiographic analysis were performed to verify the therapeutic effects of FOXA1 knockdown on bone healing. Results We identified 22 shared and differentially expressed miRNAs between hDPSC and hBMSC, 19 of which were downregulated in osteogenically induced samples. The miRNA-transcription factor interaction network showed that FOXA1 is the most significant and novel osteogenic differentiation biomarker among more than 300 transcription factors that is directly targeted by 12 miRNAs. FOXA1 knockdown significantly promoted hBMSC osteo-specific genes and increased mineral deposits in vitro. In addition, p-ERK1/2 levels were upregulated by FOXA1 silencing. Moreover, the increased osteogenic differentiation of FOXA1 knockdown hBMSCs was partially rescued by the addition of ERK1/2 signalling inhibitors. In a mouse model of femoral defects, a sheet of FOXA1-silencing BMSCs improved bone healing, as detected by microcomputed tomography and histological evaluation. Conclusion These findings collectively demonstrate that FOXA1 silencing promotes the osteogenic differentiation of BMSCs via the ERK1/2 signalling pathway, and silencing FOXA1 in vivo effectively promotes bone healing, suggesting that FOXA1 may be a novel target for bone healing.
Collapse
Affiliation(s)
- Lijun Li
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, People's Republic of China.,Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, People's Republic of China.,Clinical Research Center of Motor System Disease of Zhejiang Province, Zhejiang Province, Hangzhou City, People's Republic of China
| | - Yibo Wang
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, People's Republic of China.,Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, People's Republic of China.,Clinical Research Center of Motor System Disease of Zhejiang Province, Zhejiang Province, Hangzhou City, People's Republic of China
| | - Zhongxiang Wang
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, People's Republic of China.,Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, People's Republic of China.,Clinical Research Center of Motor System Disease of Zhejiang Province, Zhejiang Province, Hangzhou City, People's Republic of China
| | - Deting Xue
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, People's Republic of China.,Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, People's Republic of China.,Clinical Research Center of Motor System Disease of Zhejiang Province, Zhejiang Province, Hangzhou City, People's Republic of China
| | - Chengxin Dai
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, People's Republic of China.,Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, People's Republic of China.,Clinical Research Center of Motor System Disease of Zhejiang Province, Zhejiang Province, Hangzhou City, People's Republic of China
| | - Xiang Gao
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, People's Republic of China.,Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, People's Republic of China.,Clinical Research Center of Motor System Disease of Zhejiang Province, Zhejiang Province, Hangzhou City, People's Republic of China
| | - Jianfei Ma
- Key Laboratory of Image Information Processing and Intelligent Control, School of Artificial Intelligence and Automation, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Kai Hang
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, People's Republic of China. .,Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China. .,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, People's Republic of China. .,Clinical Research Center of Motor System Disease of Zhejiang Province, Zhejiang Province, Hangzhou City, People's Republic of China.
| | - Zhijun Pan
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, People's Republic of China. .,Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China. .,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, People's Republic of China. .,Clinical Research Center of Motor System Disease of Zhejiang Province, Zhejiang Province, Hangzhou City, People's Republic of China.
| |
Collapse
|
5
|
Verduci E, Calcaterra V, Di Profio E, Fiore G, Rey F, Magenes VC, Todisco CF, Carelli S, Zuccotti GV. Brown Adipose Tissue: New Challenges for Prevention of Childhood Obesity. A Narrative Review. Nutrients 2021; 13:1450. [PMID: 33923364 PMCID: PMC8145569 DOI: 10.3390/nu13051450] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/14/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
Pediatric obesity remains a challenge in modern society. Recently, research has focused on the role of the brown adipose tissue (BAT) as a potential target of intervention. In this review, we revised preclinical and clinical works on factors that may promote BAT or browning of white adipose tissue (WAT) from fetal age to adolescence. Maternal lifestyle, type of breastfeeding and healthy microbiota can affect the thermogenic activity of BAT. Environmental factors such as exposure to cold or physical activity also play a role in promoting and activating BAT. Most of the evidence is preclinical, although in clinic there is some evidence on the role of omega-3 PUFAs (EPA and DHA) supplementation on BAT activation. Clinical studies are needed to dissect the early factors and their modulation to allow proper BAT development and functions and to prevent onset of childhood obesity.
Collapse
Affiliation(s)
- Elvira Verduci
- Department of Health Sciences, University of Milan, 20146 Milan, Italy
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (V.C.); (E.D.P.); (G.F.); (V.C.M.); (C.F.T.); (G.V.Z.)
| | - Valeria Calcaterra
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (V.C.); (E.D.P.); (G.F.); (V.C.M.); (C.F.T.); (G.V.Z.)
- Pediatric and Adolescent Unit, Department of Internal Medicine, University of Pavia, 27100 Pavia, Italy
| | - Elisabetta Di Profio
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (V.C.); (E.D.P.); (G.F.); (V.C.M.); (C.F.T.); (G.V.Z.)
- Department of Animal Sciences for Health, Animal Production and Food Safety, University of Milan, 20133 Milan, Italy
| | - Giulia Fiore
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (V.C.); (E.D.P.); (G.F.); (V.C.M.); (C.F.T.); (G.V.Z.)
| | - Federica Rey
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, 20157 Milan, Italy;
- Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, University of Milan, 20157 Milan, Italy
| | - Vittoria Carlotta Magenes
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (V.C.); (E.D.P.); (G.F.); (V.C.M.); (C.F.T.); (G.V.Z.)
| | - Carolina Federica Todisco
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (V.C.); (E.D.P.); (G.F.); (V.C.M.); (C.F.T.); (G.V.Z.)
| | - Stephana Carelli
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, 20157 Milan, Italy;
- Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, University of Milan, 20157 Milan, Italy
| | - Gian Vincenzo Zuccotti
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (V.C.); (E.D.P.); (G.F.); (V.C.M.); (C.F.T.); (G.V.Z.)
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, 20157 Milan, Italy;
- Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, University of Milan, 20157 Milan, Italy
| |
Collapse
|
6
|
Nehlin JO, Jafari A, Tencerova M, Kassem M. Aging and lineage allocation changes of bone marrow skeletal (stromal) stem cells. Bone 2019; 123:265-273. [PMID: 30946971 DOI: 10.1016/j.bone.2019.03.041] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/30/2019] [Accepted: 03/31/2019] [Indexed: 01/02/2023]
Abstract
Aging is associated with decreased bone mass and accumulation of bone marrow adipocytes. Both bone forming osteoblastic cells and bone marrow adipocytes are derived from a stem cell population within the bone marrow stroma called bone marrow stromal (skeletal or mesenchymal) stem cells (BMSC). In the present review, we provide an overview, based on the current literature, regarding the physiological aging processes that cause changes in BMSC lineage allocation, enhancement of adipocyte and defective osteoblast differentiation, leading to gradual exhaustion of stem cell regenerative potential and defects in bone tissue homeostasis and metabolism. We discuss strategies to preserve the "youthful" state of BMSC, to reduce bone marrow age-associated adiposity, and to counteract the overall negative effects of aging on bone tissues with the aim of decreasing bone fragility and risk of fractures.
Collapse
Affiliation(s)
- Jan O Nehlin
- The Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology, Odense University Hospital & University of Southern Denmark, Odense, Denmark; Clinical Research Center, Copenhagen University Hospital, Hvidovre, Denmark.
| | - Abbas Jafari
- The Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology, Odense University Hospital & University of Southern Denmark, Odense, Denmark; Department of Cellular and Molecular Medicine, The Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Michaela Tencerova
- The Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology, Odense University Hospital & University of Southern Denmark, Odense, Denmark; Danish Diabetes Academy, Novo Nordisk Foundation, Odense, Denmark
| | - Moustapha Kassem
- The Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology, Odense University Hospital & University of Southern Denmark, Odense, Denmark; Department of Cellular and Molecular Medicine, The Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), Panum Institute, University of Copenhagen, Copenhagen, Denmark; Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
7
|
Jung YC, Kim HW, Min BK, Cho JY, Son HJ, Lee JY, Kim JY, Kwon SB, Li Q, Lee HW. Inhibitory Effect of Olive Leaf Extract on Obesity in High-fat Diet-induced Mice. In Vivo 2019; 33:707-715. [PMID: 31028187 PMCID: PMC6559891 DOI: 10.21873/invivo.11529] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 03/26/2019] [Accepted: 03/27/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND/AIM The rapid increase in the number of people who are overweight or obese, which increases the risk of diseases and health problems, is becoming an important issue. Herein, we investigated whether olive leaf extract (OLE) has potent anti-obesity effects in high-fat induced mouse models. MATERIALS AND METHODS C57BL/6 mice were randomized into normal control, high-fat diet (HFD), HFD with OLE, and HFD with garcinia groups and administered experimental diets for 12 weeks. Body weight and food intake were measured once per week and obesity-related biomarkers were evaluated in the serum and adipose tissue. RESULTS OLE significantly suppressed weight gain, food efficiency ratio, visceral fat accumulation, and serum lipid composition in HFD-induced mice. Furthermore, the expression of adipogenesis- and thermogenesis-related molecules was decreased in the OLE-treated group. CONCLUSION OLE prevents obesity development by regulating the expression of molecules involved in adipogenesis and thermogenesis.
Collapse
Affiliation(s)
- Yun-Chan Jung
- Institute of Research and Development, Chaon Corp., Seongnam, Republic of Korea
| | - Hyun Woo Kim
- Institute of Research and Development, Chaon Corp., Seongnam, Republic of Korea
| | - Bok Kee Min
- Nova K Health Corp., Seoul, Republic of Korea
| | | | | | | | | | | | - Qiang Li
- Institute of Research and Development, Chaon Corp., Seongnam, Republic of Korea
| | - Hee-Woo Lee
- Institute of Research and Development, Chaon Corp., Seongnam, Republic of Korea
| |
Collapse
|
8
|
Zhou J, Yang J, Wang X, Li M, Li F, Zhu E, Li X, Li X, Wang B. A Novel Regulatory Circuit "C/EBPα/miR-20a-5p/TOB2" Regulates Adipogenesis and Lipogenesis. Front Endocrinol (Lausanne) 2019; 10:894. [PMID: 31969862 PMCID: PMC6960138 DOI: 10.3389/fendo.2019.00894] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 12/05/2019] [Indexed: 12/17/2022] Open
Abstract
Recent studies have identified growing importance of microRNAs as key regulators of adipocyte differentiation. We have previously reported that miR-20a-5p is able to induce adipogenesis of established adipogenic cell lines and bone marrow derived mesenchymal stem cells (BMSCs). However, the molecular mechanisms by which miR-20a-5p controls adipogenesis and by which miR-20a-5p expression is regulated need to be further explored. In the current study we found that miR-20a-5p expression was induced during adipocyte differentiation from preadipocyte 3T3-L1 and was increased in epididymal white adipose tissue from either ob/ob mice or high fat diet-induced obese mice. Functional studies identified miR-20a-5p as a positive regulator of adipocyte differentiation and lipogenesis in 3T3-L1 by using either synthetic mimics to supplement miR-20a-5p, or using synthetic inhibitor or sponge lentivirus to inactivate endogenous miR-20a-5p. Luciferase activity assay revealed that TOB2 is a novel target of miR-20a-5p and functional experiment demonstrated its negative regulatory role in adipocyte differentiation. Moreover, Tob2 overexpression significantly attenuated adipocyte formation induced by miR-20a-5p supplementation. In-depth investigation of mechanisms that govern miR-20a-5p expression clarified that C/EBPα transcriptionally activated miR-20a-5p expression via binding to the promoter of miR-20a-5p. Taken together, we conclude that a novel C/EBPα/miR-20a-5p/TOB2 circuit exists and regulates adipogenesis and lipogenesis.
Collapse
Affiliation(s)
- Jie Zhou
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Junying Yang
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Department of Microbiology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xiaochen Wang
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Mengyue Li
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Fang Li
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Department of Microbiology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Endong Zhu
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Xuemei Li
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Xiaoxia Li
- Department of Microbiology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- *Correspondence: Xiaoxia Li
| | - Baoli Wang
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Baoli Wang
| |
Collapse
|
9
|
Gardiner JD, Abegglen LM, Huang X, Carter BE, Schackmann EA, Stucki M, Paxton CN, Lor Randall R, Amatruda JF, Putnam AR, Kovar H, Lessnick SL, Schiffman JD. C/EBPβ-1 promotes transformation and chemoresistance in Ewing sarcoma cells. Oncotarget 2018; 8:26013-26026. [PMID: 28148901 PMCID: PMC5432234 DOI: 10.18632/oncotarget.14847] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 01/16/2017] [Indexed: 12/26/2022] Open
Abstract
CEBPB copy number gain in Ewing sarcoma was previously shown to be associated with worse clinical outcome compared to tumors with normal CEBPB copy number, although the mechanism was not characterized. We employed gene knockdown and rescue assays to explore the consequences of altered CEBPB gene expression in Ewing sarcoma cell lines. Knockdown of EWS-FLI1 expression led to a decrease in expression of all three C/EBPβ isoforms while re-expression of EWS-FLI1 rescued C/EBPβ expression. Overexpression of C/EBPβ-1, the largest of the three C/EBPβ isoforms, led to a significant increase in colony formation when cells were grown in soft agar compared to empty vector transduced cells. In addition, depletion of C/EBPβ decreased colony formation, and re-expression of either C/EBPβ-1 or C/EBPβ-2 rescued the phenotype. We identified the cancer stem cell marker ALDH1A1 as a target of C/EBPβ in Ewing sarcoma. Furthermore, increased expression of C/EBPβ led to resistance to chemotherapeutic agents. In summary, we have identified CEBPB as an oncogene in Ewing sarcoma. Overexpression of C/EBPβ-1 increases transformation, upregulates expression of the cancer stem cell marker ALDH1A1, and leads to chemoresistance.
Collapse
Affiliation(s)
- Jamie D Gardiner
- Department of Pediatrics, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Lisa M Abegglen
- Department of Pediatrics, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Xiaomeng Huang
- Department of Pediatrics, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Bryce E Carter
- School of Medicine, University of Utah, Salt Lake City, UT, USA
| | | | - Marcus Stucki
- Department of Pediatrics, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Christian N Paxton
- ARUP Institute for Clinical and Experimental Pathology®, Salt Lake City, UT, USA
| | - R Lor Randall
- Department of Orthopaedic Surgery, Sarcoma Services, University of Utah, Salt Lake City, UT, USA
| | - James F Amatruda
- Department of Pediatrics, Internal Medicine and Molecular Biology, University of Texas Southwestern, Dallas, TX, USA
| | - Angelica R Putnam
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Heinrich Kovar
- Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | - Stephen L Lessnick
- Center for Childhood Cancer and Blood Disorders, Nationwide Children's Hospital, and the Division of Pediatric Heme/Onc/BMT, The Ohio State University, Columbus, OH, USA
| | - Joshua D Schiffman
- Department of Pediatrics, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
10
|
Gao Y, Li J, Xu X, Wang S, Yang Y, Zhou J, Zhang L, Zheng F, Li X, Wang B. Embelin attenuates adipogenesis and lipogenesis through activating canonical Wnt signaling and inhibits high-fat diet-induced obesity. Int J Obes (Lond) 2017; 41:729-738. [PMID: 28163317 DOI: 10.1038/ijo.2017.35] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 01/25/2017] [Accepted: 01/29/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND Recent studies suggest that Embelin, a natural plant extract might have the potential to prevent body weight gain in rats. However, the mechanisms involved remain to be elucidated. METHODS Effects of Embelin on adipocyte differentiation and lipogenesis were studied in murine ST2 stromal cells and C3H10T1/2 mesenchymal cells. The mechanisms through which Embelin regulates adipogenic differentiation and lipogenesis were explored. The in vivo anti-obesity effects of Embelin in high-fat diet (HFD)-induced obesity mice and possible transcriptional impact were investigated. RESULTS Embelin treatment suppressed ST2 and C3H10T1/2 cells to proliferate, and differentiate into mature adipocytes, along with the inhibition of adipogenic factors peroxisome proliferator-activated receptor γ, CCAAT/enhancer binding protein-α, adipocyte protein 2 and adipsin. Embelin treatment also decreased the expression levels of lipogenic factors sterol regulatory element-binding protein 1, fatty acid synthase, acetyl-CoA carboxylase 1 and stearoyl-Coenzyme A desaturase 1. Embelin promoted the translocation of β-catenin from the cytoplasm into the nucleus in C3H10T1/2. The nuclear protein levels of β-catenin and TCF-4 were increased following Embelin treatment. Furthermore, Dickkopf-1 (Dkk1) expression was downregulated by Embelin, and overexpression of Dkk1 in C3H10T1/2 reversed the inhibition of adipogenesis and lipogenesis by Embelin. In vivo studies showed that Embelin treatment reduced the gain of body weight and fat, decreased the serum level of triglycerides, free fatty acid and total cholesterol, and improved glucose tolerance and insulin resistance in HFD-fed mice. Moreover, Embelin blocked induction of adipogenic and lipogenic factors and Dkk1 in adipose tissue in HFD-fed mice. CONCLUSIONS The present work provides evidences that Embelin is effective in inhibiting adipogenesis and lipogenesis in vitro and the mechanisms may involve canonical Wnt signaling. Embelin has the potential to prevent body weight gain and fat accumulation, and to improve obesity-related glucose tolerance impairment and insulin resistance in the HFD-fed mice.
Collapse
Affiliation(s)
- Y Gao
- Key Lab of Hormones and Development (Ministry of Health), Tianjin Key Lab of Metabolic Diseases, Metabolic Diseases Hospital and Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - J Li
- Key Lab of Hormones and Development (Ministry of Health), Tianjin Key Lab of Metabolic Diseases, Metabolic Diseases Hospital and Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - X Xu
- Key Lab of Hormones and Development (Ministry of Health), Tianjin Key Lab of Metabolic Diseases, Metabolic Diseases Hospital and Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - S Wang
- Department of Microbiology, Tianjin Medical University School of Basic Medical Sciences, Tianjin, China
| | - Y Yang
- Key Lab of Hormones and Development (Ministry of Health), Tianjin Key Lab of Metabolic Diseases, Metabolic Diseases Hospital and Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - J Zhou
- Key Lab of Hormones and Development (Ministry of Health), Tianjin Key Lab of Metabolic Diseases, Metabolic Diseases Hospital and Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - L Zhang
- Department of Microbiology, Tianjin Medical University School of Basic Medical Sciences, Tianjin, China
| | - F Zheng
- Department of Biochemistry, Tianjin University of Traditional Chinese Medicine School of Integrative Medicine, Tianjin, China
| | - X Li
- Department of Microbiology, Tianjin Medical University School of Basic Medical Sciences, Tianjin, China
| | - B Wang
- Key Lab of Hormones and Development (Ministry of Health), Tianjin Key Lab of Metabolic Diseases, Metabolic Diseases Hospital and Institute of Endocrinology, Tianjin Medical University, Tianjin, China.,2011 Collaborative Innovation Center for Metabolic Diseases, Metabolic Diseases Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
11
|
Roeder E, Matthews BG, Kalajzic I. Visual reporters for study of the osteoblast lineage. Bone 2016; 92:189-195. [PMID: 27616604 PMCID: PMC5056847 DOI: 10.1016/j.bone.2016.09.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 09/06/2016] [Accepted: 09/07/2016] [Indexed: 12/24/2022]
Abstract
Advancing our understanding of osteoblast biology and differentiation is critical to elucidate the pathological mechanisms responsible for skeletal diseases such as osteoporosis. Histology and histomorphometry, the classical methods to study osteoblast biology, identify osteoblasts based on their location and morphology and ability to mineralize matrix, but do not clearly define their stage of differentiation. Introduction of visual transgenes into the cells of osteoblast lineage has revolutionized the field and resulted in a paradigm shift that allowed for specific identification and isolation of subpopulations within the osteoblast lineage. Knowledge acquired from the studies based on GFP transgenes has allowed for more precise interpretation of studies analyzing targeted overexpression or deletion of genes in the osteoblast lineage. Here, we provide a condensed overview of the currently available promoter-fluorescent reporter transgenic mice that have been generated and evaluated to varying extents. We cover different stages of the lineage as transgenes have been utilized to identify osteoprogenitors, pre-osteoblasts, osteoblasts, or osteocytes. We show that each of these promoters present with advantages and disadvantages. The studies based on the use of these reporter mice have improved our understanding of bone biology. They constitute attractive models to target osteoblasts and help to understand their cell biology.
Collapse
Affiliation(s)
- Emilie Roeder
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Brya G Matthews
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA; Department of Pathophysiology, University of Osijek, Osijek, Croatia.
| |
Collapse
|
12
|
Tencerova M, Kassem M. The Bone Marrow-Derived Stromal Cells: Commitment and Regulation of Adipogenesis. Front Endocrinol (Lausanne) 2016; 7:127. [PMID: 27708616 PMCID: PMC5030474 DOI: 10.3389/fendo.2016.00127] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 09/05/2016] [Indexed: 12/13/2022] Open
Abstract
Bone marrow (BM) microenvironment represents an important compartment of bone that regulates bone homeostasis and the balance between bone formation and bone resorption depending on the physiological needs of the organism. Abnormalities of BM microenvironmental dynamics can lead to metabolic bone diseases. BM stromal cells (also known as skeletal or mesenchymal stem cells) [bone marrow stromal stem cell (BMSC)] are multipotent stem cells located within BM stroma and give rise to osteoblasts and adipocytes. However, cellular and molecular mechanisms of BMSC lineage commitment to adipocytic lineage and regulation of BM adipocyte formation are not fully understood. In this review, we will discuss recent findings pertaining to identification and characterization of adipocyte progenitor cells in BM and the regulation of differentiation into mature adipocytes. We have also emphasized the clinical relevance of these findings.
Collapse
Affiliation(s)
- Michaela Tencerova
- Department of Molecular Endocrinology, Odense University Hospital, University of Southern Denmark, Odense, Denmark
- Danish Diabetes Academy, Novo Nordisk Foundation, Odense, Denmark
- *Correspondence: Michaela Tencerova,
| | - Moustapha Kassem
- Department of Molecular Endocrinology, Odense University Hospital, University of Southern Denmark, Odense, Denmark
- Danish Diabetes Academy, Novo Nordisk Foundation, Odense, Denmark
- Stem Cell Unit, Department of Anatomy, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
13
|
miR-140-5p regulates adipocyte differentiation by targeting transforming growth factor-β signaling. Sci Rep 2015; 5:18118. [PMID: 26657345 PMCID: PMC4676041 DOI: 10.1038/srep18118] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 11/12/2015] [Indexed: 12/21/2022] Open
Abstract
Recent emerging studies of miRNAs in adipocyte commitment provide new insights to understand the molecular basis of adipogenesis. The current study indicated that miR-140-5p was altered in primary cultured marrow stromal cells and established progenitor lines after adipogenic and/or osteogenic treatment. miR-140-5p was increased in adipose tissue in db/db obese mice vs. lean mice. Supplementing miR-140-5p activity induced stromal cell ST2 and preadipocyte 3T3-L1 to differentiate into mature adipocytes. Conversely, inhibition of the endogenous miR-140-5p repressed ST2 and 3T3-L1 to fully differentiate. By contrast, knockdown of the endogenous miR-140-5p enhanced osteoblast differentiation. Transforming growth factor-β receptor I (Tgfbr1) was shown to be a direct target of miR-140-5p. Supplementing miR-140-5p in ST2 reduced the level of TGFBR1 protein, while suppression of endogenous miR-140-5p increased TGFBR1. Overexpression of Tgfbr1 inhibited, whereas knockdown of Tgfbr1 promoted adipogenic differentiation of ST2 cells. Further investigation of mechanisms that control miR-140-5p expression revealed that C/EBPα induced transcriptional activity of the miR-140-5p promoter. Removal of the putative response element of C/EBP from the promoter abolished the enhancement of the promoter activity by C/EBPα, suggesting that C/EBPα transcriptionally controls miR-140-5p expression. Taken together, our study provides evidences that miR-140-5p regulates adipocyte differentiation through a C/EBP/miR-140-5p/TGFBR1 regulatory feedback loop.
Collapse
|
14
|
Garcia-Gomez A, De Las Rivas J, Ocio EM, Díaz-Rodríguez E, Montero JC, Martín M, Blanco JF, Sanchez-Guijo FM, Pandiella A, San Miguel JF, Garayoa M. Transcriptomic profile induced in bone marrow mesenchymal stromal cells after interaction with multiple myeloma cells: implications in myeloma progression and myeloma bone disease. Oncotarget 2015; 5:8284-305. [PMID: 25268740 PMCID: PMC4226683 DOI: 10.18632/oncotarget.2058] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Despite evidence about the implication of the bone marrow (BM) stromal microenvironment in multiple myeloma (MM) cell growth and survival, little is known about the effects of myelomatous cells on BM stromal cells. Mesenchymal stromal cells (MSCs) from healthy donors (dMSCs) or myeloma patients (pMSCs) were co-cultured with the myeloma cell line MM.1S, and the transcriptomic profile of MSCs induced by this interaction was analyzed. Deregulated genes after co-culture common to both d/pMSCs revealed functional involvement in tumor microenvironment cross-talk, myeloma growth induction and drug resistance, angiogenesis and signals for osteoclast activation and osteoblast inhibition. Additional genes induced by co-culture were exclusively deregulated in pMSCs and predominantly associated to RNA processing, the ubiquitine-proteasome pathway, cell cycle regulation, cellular stress and non-canonical Wnt signaling. The upregulated expression of five genes after co-culture (CXCL1, CXCL5 and CXCL6 in d/pMSCs, and Neuregulin 3 and Norrie disease protein exclusively in pMSCs) was confirmed, and functional in vitro assays revealed putative roles in MM pathophysiology. The transcriptomic profile of pMSCs co-cultured with myeloma cells may better reflect that of MSCs in the BM of myeloma patients, and provides new molecular insights to the contribution of these cells to MM pathophysiology and to myeloma bone disease.
Collapse
Affiliation(s)
- Antonio Garcia-Gomez
- Centro de Investigación del Cáncer, IBMCC (Universidad de Salamanca-CSIC), Salamanca, Spain. Hospital Universitario de Salamanca-IBSAL, Salamanca, Spain. Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Salamanca, Spain
| | - Javier De Las Rivas
- Centro de Investigación del Cáncer, IBMCC (Universidad de Salamanca-CSIC), Salamanca, Spain
| | - Enrique M Ocio
- Centro de Investigación del Cáncer, IBMCC (Universidad de Salamanca-CSIC), Salamanca, Spain. Hospital Universitario de Salamanca-IBSAL, Salamanca, Spain
| | - Elena Díaz-Rodríguez
- Centro de Investigación del Cáncer, IBMCC (Universidad de Salamanca-CSIC), Salamanca, Spain
| | - Juan C Montero
- Centro de Investigación del Cáncer, IBMCC (Universidad de Salamanca-CSIC), Salamanca, Spain
| | - Montserrat Martín
- Centro de Investigación del Cáncer, IBMCC (Universidad de Salamanca-CSIC), Salamanca, Spain. Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Salamanca, Spain
| | - Juan F Blanco
- Hospital Universitario de Salamanca-IBSAL, Salamanca, Spain
| | - Fermín M Sanchez-Guijo
- Hospital Universitario de Salamanca-IBSAL, Salamanca, Spain. Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Salamanca, Spain
| | - Atanasio Pandiella
- Centro de Investigación del Cáncer, IBMCC (Universidad de Salamanca-CSIC), Salamanca, Spain. Hospital Universitario de Salamanca-IBSAL, Salamanca, Spain
| | - Jesús F San Miguel
- Centro de Investigación del Cáncer, IBMCC (Universidad de Salamanca-CSIC), Salamanca, Spain. Hospital Universitario de Salamanca-IBSAL, Salamanca, Spain. Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Salamanca, Spain
| | - Mercedes Garayoa
- Centro de Investigación del Cáncer, IBMCC (Universidad de Salamanca-CSIC), Salamanca, Spain. Hospital Universitario de Salamanca-IBSAL, Salamanca, Spain. Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Salamanca, Spain
| |
Collapse
|
15
|
Guan X, Gao Y, Zhou J, Wang J, Zheng F, Guo F, Chang A, Li X, Wang B. miR-223 Regulates Adipogenic and Osteogenic Differentiation of Mesenchymal Stem Cells Through a C/EBPs/miR-223/FGFR2 Regulatory Feedback Loop. Stem Cells 2015; 33:1589-600. [PMID: 25641499 DOI: 10.1002/stem.1947] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 12/07/2014] [Indexed: 01/10/2023]
Affiliation(s)
- Xiaohui Guan
- Key Laboratory of Hormones and Development (Ministry of Health); Metabolic Diseases Hospital & Institute of Endocrinology; and Tianjin Medical University; Tianjin People's Republic of China
| | - Yifei Gao
- Key Laboratory of Hormones and Development (Ministry of Health); Metabolic Diseases Hospital & Institute of Endocrinology; and Tianjin Medical University; Tianjin People's Republic of China
| | - Jie Zhou
- Key Laboratory of Hormones and Development (Ministry of Health); Metabolic Diseases Hospital & Institute of Endocrinology; and Tianjin Medical University; Tianjin People's Republic of China
| | - Jun Wang
- Key Laboratory of Hormones and Development (Ministry of Health); Metabolic Diseases Hospital & Institute of Endocrinology; and Tianjin Medical University; Tianjin People's Republic of China
| | - Fang Zheng
- College of Basic Medical Sciences; Tianjin Traditional Medical University; Tianjin People's Republic of China
| | - Fei Guo
- Key Laboratory of Hormones and Development (Ministry of Health); Metabolic Diseases Hospital & Institute of Endocrinology; and Tianjin Medical University; Tianjin People's Republic of China
| | - Ailing Chang
- Key Laboratory of Hormones and Development (Ministry of Health); Metabolic Diseases Hospital & Institute of Endocrinology; and Tianjin Medical University; Tianjin People's Republic of China
| | - Xiaoxia Li
- College of Basic Medical Sciences; Tianjin Medical University; Tianjin People's Republic of China
| | - Baoli Wang
- Key Laboratory of Hormones and Development (Ministry of Health); Metabolic Diseases Hospital & Institute of Endocrinology; and Tianjin Medical University; Tianjin People's Republic of China
| |
Collapse
|
16
|
miR-20a regulates adipocyte differentiation by targeting lysine-specific demethylase 6b and transforming growth factor-β signaling. Int J Obes (Lond) 2015; 39:1282-91. [PMID: 25817070 DOI: 10.1038/ijo.2015.43] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 03/10/2015] [Accepted: 03/21/2015] [Indexed: 12/26/2022]
Abstract
BACKGROUND Several types of microRNAs (miRNAs) have recently been defined as important regulators in adipocyte differentiation, the role of other miRNAs in the processes and the mechanisms involved remain to be explored. METHODS miR-20a expression was quantified in primary cultured marrow stromal cells and adipogenic cell lines after adipogenic treatment. Effects of miR-20a on adipocyte differentiation were studied following supplementing or depleting miR-20a in murine 3T3-L1 preadipocytes, ST2 stromal cells and C3H10T1/2 mesenchymal cells. Bioinformatics prediction of miRNA targets was performed, and potential targets of miR-20a were verified by using dual luciferase activity assays. Gain-of-function and loss-of-function studies were performed to examine the effects of the target genes on adipocyte differentiation. RESULTS miR-20a was induced in primary cultured marrow stromal cells and established adipogenic lines after adipogenic treatment. Supplementing miR-20a activity suppressed the growth of 3T3-L1 preadipocytes and induced 3T3-L1, ST2 and C3H10T1/2 cells to differentiate into mature adipocytes, along with the induction of adipocyte-specific transcription factors peroxisome proliferator-activated receptor γ (PPARγ), CCAAT/enhancer binding protein-α (C/EBPα), C/EBPβ and the marker gene adipocyte protein 2 (aP2). Conversely, inhibition of the endogenous miR-20a repressed 3T3-L1, ST2 and C3H10T1/2 cells to fully differentiate. Transforming growth factor-β receptor II (Tgfbr2) and lysine-specific demethylase 6b (Kdm6b) were shown to be direct targets of miR-20a. Supplementing miR-20a activity in ST2 reduced levels of KDM6B and TGFBR2 proteins, while suppression of endogenous miR-20a increased KDM6B and TGFBR2. While TGF-β signaling is a well-documented inhibitor of adipogenesis, the effects of Kdm6b on adipocyte formation need to be clarified. We demonstrated that overexpression of Kdm6b inhibited, while knockdown of Kdm6b promoted the differentiation of the ST2 cells into mature adipocytes. CONCLUSION The present work provides evidence that mouse miR-20a promotes adipocyte progenitor cells to differentiate and this function may depend upon its inhibitory effects on Kdm6b and TGF-β signaling.
Collapse
|
17
|
Meyer MB, Benkusky NA, Pike JW. Selective Distal Enhancer Control of the Mmp13 Gene Identified through Clustered Regularly Interspaced Short Palindromic Repeat (CRISPR) Genomic Deletions. J Biol Chem 2015; 290:11093-107. [PMID: 25773540 DOI: 10.1074/jbc.m115.648394] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Indexed: 12/29/2022] Open
Abstract
Matrix metalloproteinase 13 (Mmp13, collagenase-3) plays an essential role in bone metabolism and mineral homeostasis. It is regulated by numerous factors, including BMP-2, parathyroid hormone, and 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3), through transcription factors such as Runt-related transcription factor 2 (RUNX2), CCAAT/enhancer-binding protein β (C/EBPβ), OSX, and vitamin D receptor (VDR). During osteoblast maturation, the basal expression of Mmp13 and its sensitivity to 1,25(OH)2D3 are strikingly increased. In this report, ChIP-sequencing analysis in mouse preosteoblasts revealed that the Mmp13 gene was probably regulated by three major enhancers located -10, -20, and -30 kb upstream of the gene promoter, occupied by activated VDR and prebound C/EBPβ and RUNX2, respectively. Initially, bacterial artificial chromosome clone recombineering and traditional mutagenesis defined binding sites for VDR and RUNX2. We then employed a CRISPR/Cas9 gene editing approach to delete the -10 and -30 kb Mmp13 enhancers, a region proximal to the promoter, and VDR or RUNX2. VDR-mediated up-regulation of Mmp13 transcription was completely abrogated upon removal of the -10 kb enhancer, resulting in a 1,25(OH)2D3-directed repression of Mmp13. Deletion of either the -30 kb enhancer or RUNX2 resulted in a complete loss of basal transcript activity and a ChIP-identified destabilization of the chromatin enhancer environment and factor binding. Whereas enhancer deletions only affected Mmp13 expression, the RUNX2 deletion led to changes in gene expression, a reduction in cellular proliferation, and an inability to differentiate. We conclude that the Mmp13 gene is regulated via at least three specific distal enhancers that display independent activities yet are able to integrate response from multiple signaling pathways in a model of activation and suppression.
Collapse
Affiliation(s)
- Mark B Meyer
- From the Department of Biochemistry, University of Wisconsin, Madison, Wisconsin 53706
| | - Nancy A Benkusky
- From the Department of Biochemistry, University of Wisconsin, Madison, Wisconsin 53706
| | - J Wesley Pike
- From the Department of Biochemistry, University of Wisconsin, Madison, Wisconsin 53706
| |
Collapse
|
18
|
Brommage R, Liu J, Hansen GM, Kirkpatrick LL, Potter DG, Sands AT, Zambrowicz B, Powell DR, Vogel P. High-throughput screening of mouse gene knockouts identifies established and novel skeletal phenotypes. Bone Res 2014; 2:14034. [PMID: 26273529 PMCID: PMC4472125 DOI: 10.1038/boneres.2014.34] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 07/29/2014] [Accepted: 07/31/2014] [Indexed: 12/13/2022] Open
Abstract
Screening gene function in vivo is a powerful approach to discover novel drug targets. We present high-throughput screening (HTS) data for 3 762 distinct global gene knockout (KO) mouse lines with viable adult homozygous mice generated using either gene-trap or homologous recombination technologies. Bone mass was determined from DEXA scans of male and female mice at 14 weeks of age and by microCT analyses of bones from male mice at 16 weeks of age. Wild-type (WT) cagemates/littermates were examined for each gene KO. Lethality was observed in an additional 850 KO lines. Since primary HTS are susceptible to false positive findings, additional cohorts of mice from KO lines with intriguing HTS bone data were examined. Aging, ovariectomy, histomorphometry and bone strength studies were performed and possible non-skeletal phenotypes were explored. Together, these screens identified multiple genes affecting bone mass: 23 previously reported genes (Calcr, Cebpb, Crtap, Dcstamp, Dkk1, Duoxa2, Enpp1, Fgf23, Kiss1/Kiss1r, Kl (Klotho), Lrp5, Mstn, Neo1, Npr2, Ostm1, Postn, Sfrp4, Slc30a5, Slc39a13, Sost, Sumf1, Src, Wnt10b), five novel genes extensively characterized (Cldn18, Fam20c, Lrrk1, Sgpl1, Wnt16), five novel genes with preliminary characterization (Agpat2, Rassf5, Slc10a7, Slc26a7, Slc30a10) and three novel undisclosed genes coding for potential osteoporosis drug targets.
Collapse
Affiliation(s)
| | - Jeff Liu
- Lexicon Pharmaceuticals , The Woodlands, TX, USA
| | | | | | | | | | | | | | - Peter Vogel
- Lexicon Pharmaceuticals , The Woodlands, TX, USA
| |
Collapse
|
19
|
Meyer MB, Benkusky NA, Pike JW. The RUNX2 cistrome in osteoblasts: characterization, down-regulation following differentiation, and relationship to gene expression. J Biol Chem 2014; 289:16016-31. [PMID: 24764292 PMCID: PMC4047377 DOI: 10.1074/jbc.m114.552216] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 04/23/2014] [Indexed: 01/09/2023] Open
Abstract
RUNX2 is a transcription factor that is first expressed in early osteoblast-lineage cells and represents a primary determinant of osteoblastogenesis. While numerous target genes are regulated by RUNX2, little is known of sites on the genome occupied by RUNX2 or of the gene networks that are controlled by these sites. To explore this, we conducted a genome-wide analysis of the RUNX2 cistrome in both pre-osteoblastic MC3T3-E1 cells (POB) and their mature osteoblast progeny (OB), characterized the two cistromes and assessed their relationship to changes in gene expression. We found that although RUNX2 was widely bound to the genome in POB cells, this binding profile was reduced upon differentiation to OBs. Numerous sites were lost upon differentiation, new sites were also gained; many sites remained common to both cell states. Additional features were identified as well including location relative to potential target genes, abundance with respect to single genes, the frequent presence of a consensus TGTGGT RUNX2 binding motif, co-occupancy by C/EBPβ and the presence of a typical epigenetic histone enhancer signature. This signature was changed quantitatively following differentiation. While RUNX2 binding sites were associated extensively with adjacent genes, the distal nature of the majority of these sites prevented assessment of whether they represented direct targets of RUNX2 action. Changes in gene expression, however, revealed an abundance of genes that contained RUNX2 binding sites and were regulated in concert. These studies establish a basis for further analysis of the role of RUNX2 activity and its function during osteoblast lineage maturation.
Collapse
Affiliation(s)
- Mark B Meyer
- From the Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - Nancy A Benkusky
- From the Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - J Wesley Pike
- From the Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706
| |
Collapse
|
20
|
Zanotti S, Kalajzic I, Aguila HL, Canalis E. Sex and genetic factors determine osteoblastic differentiation potential of murine bone marrow stromal cells. PLoS One 2014; 9:e86757. [PMID: 24489784 PMCID: PMC3904935 DOI: 10.1371/journal.pone.0086757] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 12/16/2013] [Indexed: 01/08/2023] Open
Abstract
Sex and genetic factors determine skeletal mass, and we tested whether bone histomorphometric parameters were sexually dimorphic in femurs from 1 to 6 month old C57BL/6 mice. Trabecular bone volume declined more rapidly in female mice than in male littermates because of enhanced bone resorption. Although bone formation was not different between sexes, female mice exhibited a higher number of osteoblasts than male littermates, suggesting that osteoblasts from female mice may have a reduced ability to form bone. To determine the impact of sex on osteoblastogenesis, we investigated the potential for osteoblastic differentiation of bone marrow stromal cells from C57BL/6, Friend leukemia virus-B (FVB), C3H/HeJ and BALB/c mice of both sexes. Bone marrow stromal cells from female FVB, C57BL/6 and C3H/HeJ mice exhibited lower Alpl and Osteocalcin expression and alkaline phosphatase activity, and formed fewer mineralized nodules than cells from male littermates. Proliferative capacity was greater in cells from male than female C57BL/6, but not FVB, mice. Sorting of bone marrow stromal cells from mice expressing an α-Smooth muscle actin-green fluorescent protein transgene, revealed a higher yield of mesenchymal stem cells in cultures from male mice than in those from female littermates. Sex had a modest impact on osteoblastic differentiation of mesenchymal stem cells. To determine the influence of sex and genetic factors on osteoblast function, calvarial osteoblasts were harvested from C57BL/6, FVB, C3H/HeJ and BALB/c mice. Alpl expression and activity were lower in osteoblasts from C57BL/6 and C3H/HeJ, but not FVB or BALB/c, female mice than in cells from littermates. Sex had no effect on osteoclastogenesis of bone marrow cultures of C57BL/6 mice, but osteoblasts from female mice exhibited higher Rankl and lower Opg expression than cells from male littermates. In conclusion, osteoblastogenesis is sexually dimorphic and influenced by genetic factors.
Collapse
Affiliation(s)
- Stefano Zanotti
- Department of Research, Saint Francis Hospital and Medical Center, Hartford, Connecticut, United States of America
- University of Connecticut School of Medicine, University of Connecticut Health Center, Farmington, Connecticut, United States of America
- * E-mail:
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Hector Leonardo Aguila
- Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Ernesto Canalis
- Department of Research, Saint Francis Hospital and Medical Center, Hartford, Connecticut, United States of America
- University of Connecticut School of Medicine, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| |
Collapse
|
21
|
Wang J, Guan X, Guo F, Zhou J, Chang A, Sun B, Cai Y, Ma Z, Dai C, Li X, Wang B. miR-30e reciprocally regulates the differentiation of adipocytes and osteoblasts by directly targeting low-density lipoprotein receptor-related protein 6. Cell Death Dis 2013; 4:e845. [PMID: 24113179 PMCID: PMC3824666 DOI: 10.1038/cddis.2013.356] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 08/09/2013] [Accepted: 08/12/2013] [Indexed: 01/08/2023]
Abstract
Reciprocal relationship usually exists between osteoblastogenesis and adipogenesis, with factors stimulating one of these processes at the same time inhibiting the other. In the present study, miR-30e was found to be involved in the reciprocal regulation of osteoblast and adipocyte differentiation. Our data indicated that miR-30e was induced in primarily cultured mouse bone marrow stromal cell, mesenchymal cell line C3H10T1/2 and preadipocyte 3T3-L1 after adipogenic treatment. Conversely, it was reduced in mouse stromal line ST2 and preosteoblast MC3T3-E1 after osteogenic treatment. Enforced expression of miR-30e in 3T3-L1 significantly suppressed the growth of the cells and induced the preadipocytes to differentiate into mature adipocytes, along with increased expression of adipocyte-specific transcription factors peroxisome proliferator-activated receptor-γ (PPARγ), CCAAT/enhancer binding protein-α (C/EBPα) and C/EBPβ, and the marker gene aP2. In contrast, inhibition of the endogenous miR-30e enhanced the cell growth and repressed preadipocytes to differentiate. Conversely, supplementing miR-30e activity blocked, whereas knocking down miR-30e enforced the preosteoblast MC3T3-E1 to fully differentiate. Furthermore, miR-30e overexpression stimulated adipocyte formation and inhibited osteoblast differentiation from marrow stromal cells. Low-density lipoprotein receptor-related protein 6 (LRP6), one of the critical coreceptor for Wnts, was shown to be a direct target of miR-30e by using the luciferase assay. Knockdown of LRP6 in 3T3-L1 cells downregulated β-catenin/T-cell factor (TCF) transcriptional activity and dramatically potentiated the differentiation of the cells into mature adipocytes. Taken together, the present work suggests that the expression of miR-30e is indispensable for maintaining the balance of adipocytes and osteoblasts by targeting the canonical Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- J Wang
- Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Institute of Endocrinology, Tianjin Medical University, Tianjin 300070, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Ponrartana S, Hu HH, Gilsanz V. On the relevance of brown adipose tissue in children. Ann N Y Acad Sci 2013; 1302:24-29. [DOI: 10.1111/nyas.12195] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
| | | | - Vicente Gilsanz
- Departments of Radiology; Los Angeles California
- Pediatrics, Children's Hospital Los Angeles; Los Angeles California
| |
Collapse
|
23
|
Lee P, Brychta RJ, Collins MT, Linderman J, Smith S, Herscovitch P, Millo C, Chen KY, Celi FS. Cold-activated brown adipose tissue is an independent predictor of higher bone mineral density in women. Osteoporos Int 2013; 24:1513-8. [PMID: 22890364 PMCID: PMC5572572 DOI: 10.1007/s00198-012-2110-y] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 07/23/2012] [Indexed: 12/14/2022]
Abstract
UNLABELLED In animals, defective brown adipogenesis leads to bone loss. Whether brown adipose tissue (BAT) mass relates to bone mineral density (BMD) in humans is unclear. We determined the relationship between BAT mass and BMD by cold-stimulated positron-emission tomography (PET) and dual-energy X-ray absorptiometry (DXA) in healthy volunteers. Higher BAT mass was associated with higher BMD in healthy women, but not in men, independent of age and body composition. INTRODUCTION Contrary to the traditional belief that BAT is present only in infants, recent studies revealed significant depots of BAT present in adult humans. In animals, defective brown adipogenesis leads to bone loss. While white adipose tissue mass is a known determinant of BMD in humans, the relationship between BAT and BMD in humans is unclear. We thus examined the relationship between BAT and BMD in healthy adults. METHODS BAT volume (ml) and activity (standard uptake value) were determined by 18F-fluorodeoxyglucose PET after overnight mild cold exposure at 19 °C, and BMD was determined by DXA. RESULTS Among 24 healthy adults (age 28±1 years, F=10), BAT volumes were 82.4±99.5 ml in women and 49.7±54.5 ml in men. Women manifested significantly higher BAT activity, by 9.4±8.1% (p=0.03), than men. BAT volume correlated positively with total and spine BMD (r2=0.40 and 0.49, respectively, p<0.02) in women and remained a significant predictor after adjustment for age, fat, and lean body mass (p<0.05). Total and spine BMD were higher in women who harbored visually detectable BAT on PET images than those without by 11±2% (p=0.02) and 22±2% (p<0.01), respectively. No associations were observed between BAT parameters and BMD in men. CONCLUSIONS This study demonstrated higher BMD among healthy women with more abundant BAT, independent of age and other body compositional parameters. This was not observed in men. The data suggest that brown adipogenesis may be physiologically related to modulation of bone density.
Collapse
Affiliation(s)
- P Lee
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bldg 10, CRC, 10 Center Drive, Bethesda, MD, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Bredella MA, Fazeli PK, Lecka-Czernik B, Rosen CJ, Klibanski A. IGFBP-2 is a negative predictor of cold-induced brown fat and bone mineral density in young non-obese women. Bone 2013; 53:336-9. [PMID: 23291564 PMCID: PMC3594375 DOI: 10.1016/j.bone.2012.12.046] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 12/18/2012] [Accepted: 12/19/2012] [Indexed: 11/19/2022]
Abstract
Recent studies have shown a positive correlation between brown adipose tissue (BAT) and bone mineral density (BMD). However, mechanisms underlying this relationship are unknown. Insulin-like growth factor 1 (IGF-1) is an important regulator of stem cell differentiation promoting bone formation. IGF binding protein 2 (IGFBP-2) binds IGF-1 in the circulation and has been reported to inhibit bone formation in humans. IGF-1 is also a crucial regulator of brown adipocyte differentiation. We hypothesized that IGFBP-2 is a negative and IGF-1 a positive regulator of BAT-mediated osteoblastogenesis. We therefore investigated a cohort of 15 women (mean age 27.7±5.7years): 5 with anorexia nervosa (AN) in whom IGF-1 levels were low due to starvation, 5 recovered AN (AN-R), and 5 women of normal weight. All subjects underwent assessment of cold-activated BAT by PET/CT, BMD of the spine, hip, femoral neck, and total body by DXA, thigh muscle area by MRI, IGF-1 and IGFBP-2. There was a positive correlation between BAT and BMD and an inverse association between IGFBP-2 and both BAT and BMD. There was no association between IGF-1 and BAT. We show for the first time that IGFBP-2 is a negative predictor of cold-induced BAT and BMD in young non-obese women, suggesting that IGFBP-2 may serve as a regulator of BAT-mediated osteoblastogenesis.
Collapse
Affiliation(s)
- Miriam A Bredella
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| | | | | | | | | |
Collapse
|
25
|
Abstract
Brown adipose tissue (BAT) was thought to disappear after infancy. Recent findings of BAT in patients undergoing positron emission tomography/computed tomography (PET/CT) have renewed the interest in deciphering the relevance of this tissue in humans. Available data suggest that BAT is more prevalent in children than in adults and that its activation during adolescence is associated with significantly lower gains in weight and adiposity. Data also show that pediatric patients with metabolically active BAT on PET/CT examinations have significantly greater muscle volume than patients without identifiable BAT. Both the activity and the amount of BAT increase during puberty. The magnitude of the increase is higher in boys as compared with girls and is closely related to gains in muscle volume. Hence, concurrent with the gains in skeletal muscle during infancy and puberty, all infants and adolescents accumulate large amounts of BAT. These observations are consistent with in vitro investigations suggesting close interactions between brown adipocytes, white adipocytes, and myocytes. In this review, we discuss the potential role of this tissue in regulating weight and musculoskeletal development in children.
Collapse
Affiliation(s)
- Vicente Gilsanz
- Department of Radiology, Children's Hospital Los Angeles, Los Angeles, CA, USA.
| | - Houchun H. Hu
- Department of Radiology, Children’s Hospital Los Angeles, Los Angeles, CA 90027
,Department of Electrical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90027
| | - Shingo Kajimura
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143
,Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143
| |
Collapse
|
26
|
Reddi D, Belibasakis GN. Transcriptional profiling of bone marrow stromal cells in response to Porphyromonas gingivalis secreted products. PLoS One 2012; 7:e43899. [PMID: 22937121 PMCID: PMC3427182 DOI: 10.1371/journal.pone.0043899] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2012] [Accepted: 07/30/2012] [Indexed: 12/18/2022] Open
Abstract
Periodontitis is an infectious inflammatory disease that destroys the tooth-supporting (periodontal) tissues. Porphyromonas gingivalis is an oral pathogen highly implicated in the pathogenesis of this disease. It can exert its effects to a number of cells, including osteogenic bone marrow stromal cells which are important for homeostastic capacity of the tissues. By employing gene microarray technology, this study aimed to describe the overall transcriptional events (>2-fold regulation) elicited by P. gingivalis secreted products in bone marrow stromal cells, and to dissect further the categories of genes involved in bone metabolism, inflammatory and immune responses. After 6 h of challenge with P. gingivalis, 271 genes were up-regulated whereas 209 genes were down-regulated, whereas after 24 h, these numbers were 259 and 109, respectively. The early (6 h) response was characterised by regulation of genes associated with inhibition of cell cycle, induction of apoptosis and loss of structural integrity, whereas the late (24 h) response was characterised by induction of chemokines, cytokines and their associated intracellular pathways (such as NF-κB), mediators of connective tissue and bone destruction, and suppression of regulators of osteogenic differentiation. The most strongly up-regulated genes were lipocalin 2 (LCN2) and serum amyloid A3 (SAA3), both encoding for proteins of the acute phase inflammatory response. Collectively, these transcriptional changes elicited by P. gingivalis denote that the fundamental cellular functions are hindered, and that the cells acquire a phenotype commensurate with propagated innate immune response and inflammatory-mediated tissue destruction. In conclusion, the global transcriptional profile of bone marrow stromal cells in response to P. gingivalis is marked by deregulated homeostatic functions, with implications in the pathogenesis of periodontitis.
Collapse
Affiliation(s)
- Durga Reddi
- Centre for Adult Oral Health, Barts and the London Institute of Dentistry, Queen Mary University of London, London, United Kingdom
| | - Georgios N. Belibasakis
- Centre for Adult Oral Health, Barts and the London Institute of Dentistry, Queen Mary University of London, London, United Kingdom
- Oral Microbiology and Immunology, Institute of Oral Biology, Center of Dental Medicine, University of Zürich, Zürich, Switzerland
- * E-mail:
| |
Collapse
|
27
|
Ponrartana S, Aggabao PC, Hu HH, Aldrovandi GM, Wren TAL, Gilsanz V. Brown adipose tissue and its relationship to bone structure in pediatric patients. J Clin Endocrinol Metab 2012; 97:2693-8. [PMID: 22593587 PMCID: PMC3410267 DOI: 10.1210/jc.2012-1589] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
CONTEXT Emerging evidence suggests a possible link between brown adipose tissue (BAT) and bone metabolism. OBJECTIVE The objective of this study was to examine the relationships between BAT and bone cross-sectional dimensions in children and adolescents. DESIGN This was a cross-sectional study. SETTING The study was conducted at a pediatric referral center. PATIENTS Patients included 40 children and teenagers (21 males and 19 females) successfully treated for pediatric malignancies. INTERVENTIONS There were no interventions. MAIN OUTCOME MEASURES The volume of BAT was determined by fluorodeoxyglucose-positron emission tomography/computed tomography. Measures of the cross-sectional area and cortical bone area and measures of thigh musculature and sc fat were determined at the midshaft of the femur. RESULTS Regardless of sex, there were significant correlations seen between BAT volume and the cross-sectional dimensions of the bone (r values between 0.68 and 0.77; all P ≤ 0 .001). Multiple regression analyses indicated that the volume of BAT predicted femoral cross-sectional area and cortical bone area, even after accounting for height, weight, and gender. The addition of muscle as an independent variable increased the predictive power of the model but significantly decreased the contribution of BAT. CONCLUSIONS The volume of BAT is positively associated with the amount of bone and the cross-sectional size of the femur in children and adolescents. This relation between BAT and bone structure could, at least in part, be mediated by muscle.
Collapse
Affiliation(s)
- Skorn Ponrartana
- Department of Radiology, Viterbi School of Engineering, University of Southern California, Los Angeles, California 90027, USA
| | | | | | | | | | | |
Collapse
|
28
|
Bredella MA, Fazeli PK, Freedman LM, Calder G, Lee H, Rosen CJ, Klibanski A. Young women with cold-activated brown adipose tissue have higher bone mineral density and lower Pref-1 than women without brown adipose tissue: a study in women with anorexia nervosa, women recovered from anorexia nervosa, and normal-weight women. J Clin Endocrinol Metab 2012; 97:E584-90. [PMID: 22259053 PMCID: PMC3319179 DOI: 10.1210/jc.2011-2246] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
CONTEXT Anorexia nervosa (AN) is associated with depletion of body fat, loss of bone mineral density (BMD), and impaired thermogenesis. Brown adipose tissue (BAT) is lower in obese individuals and decreases during aging. Recent studies have suggested a link between BAT and bone metabolism. OBJECTIVE Our objective was to investigate the presence and quantity of BAT in patients with AN, recovered AN (AN-R), and normal-weight controls and to study the relationship between BAT and BMD and body composition and investigate hormonal predictors of BAT. DESIGN AND SETTING This was a cross-sectional study at a clinical research center. PATIENTS Patients included 15 women: five with AN (mean age 30 ± 6.3 yr), five AN-R, and five healthy nonobese controls of comparable age. MAIN OUTCOME MEASURES Cold-activated BAT was determined by fluorodeoxyglucose-positron emission tomography/computed tomography. BMD of total-body, spine, and hip, fat and lean mass was determined by dual-energy x-ray absorptiometry. Single-slice magnetic resonance imaging at L4 was done for abdominal fat compartments, and preadipocyte factor-1 (Pref-1), T₃, and T₄ were measured. RESULTS Within the AN group, one of five; in the AN-R group, two of five; and in the healthy nonobese control group, four of five subjects were BAT positive. Subjects were divided into groups based on the presence (n = 7) or absence (n = 8) of BAT. Both groups were of comparable age and body mass index. Women with BAT had higher total-body BMD, higher T₃, and lower Pref-1 compared with women without BAT. There was a positive correlation between BAT and BMD that remained significant after controlling for disease status and body mass index. CONCLUSION Young women with AN have low cold-activated BAT, which may be due to impaired BAT thermogenesis. Young women with BAT have higher BMD and lower Pref-1 compared with women without BAT, suggesting that BAT may be involved in the regulation of stem cell differentiation into the bone lineage at the expense of adipogenesis.
Collapse
Affiliation(s)
- Miriam A Bredella
- Department of Radiology, Massachusetts General Hospital, Yawkey 6E, 55 Fruit Street, Boston, Massachusetts 02114, USA.
| | | | | | | | | | | | | |
Collapse
|
29
|
Williams PJ, Nishu K, Rahman MM. HDAC inhibitor trichostatin A suppresses osteoclastogenesis by upregulating the expression of C/EBP-β and MKP-1. Ann N Y Acad Sci 2012; 1240:18-25. [PMID: 22172035 DOI: 10.1111/j.1749-6632.2011.06286.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Histone deacetylases (HDACs) remove the acetyl groups from the lysine residues of histone tails, leading to the formation of a condensed and transcriptionally silenced chromatin. HDAC inhibitors (HDACi) block this action and can result in hyperacetylation of histones, leading to a less compact and more transcriptionally active chromatin and thereby, gene expression. Previously, we have shown that HDACi inhibit osteoclast differentiation. However, which genes are transcriptionally activated following hyperacetylation of histones, and lead to the suppression of osteoclastogenesis, has yet to be elucidated. In this study, we show that an HDACi, trichostatin A (TSA), inhibits the receptor activator of the nuclear factor-κB (NF-κB) ligand (RANKL)-stimulated TNF-α production, NF-κB activation, and bone resorbing pit formation, and downregulates c-Fos and NFATc1 in RAW 264.7 cells. Interestingly, expression of antiosteoclastogenic factors CCAAT enhancer binding protein (C/EBP)-β and mitogen-activated protein kinase phosphatase (MKP)-1 was significantly upregulated in TSA-treated, RANKL-stimulated RAW 264.7 cells. These findings suggest that TSA upregulates the expression of C/EBP-β and MKP-1, which may downregulate pro-osteoclastogenic factors and signaling molecules, ultimately suppressing osteoclastogenesis.
Collapse
Affiliation(s)
- Paul J Williams
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | | | | |
Collapse
|
30
|
Smink JJ, Leutz A. Instruction of mesenchymal cell fate by the transcription factor C/EBPβ. Gene 2012; 497:10-7. [PMID: 22306325 DOI: 10.1016/j.gene.2012.01.043] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Revised: 12/13/2011] [Accepted: 01/19/2012] [Indexed: 01/10/2023]
Abstract
The transcription factor CCAAT/enhancer binding protein beta (C/EBPβ) plays a role in the differentiation of a large variety of cell types. C/EBPβ was initially described as an early inducer of adipocyte differentiation, however, recent data have shown that this is not the only mesenchymal cell lineage where C/EBPβ has an instructive function. Mouse models and tissue culture studies have now established a regulatory role of C/EBPβ in osteoblast and in chondrocyte differentiation. These three different cell lineages are derived from the same precursor, the mesenchymal stem cell (MSC). This review will focus on the emerging role of C/EBPβ and its different protein isoforms in various mesenchymal cell lineages and its function in adipocyte, chondrocyte and osteoblast differentiation. Moreover, the mesenchymal stem cell has attracted the attention of regenerative medicine in recent years, and the possible role of C/EBPβ in this respect will be discussed.
Collapse
Affiliation(s)
- Jeske J Smink
- Max Delbrueck Center for Molecular Medicine, Berlin-Brandenburg Center for Regenerative Therapies, Berlin, Germany
| | | |
Collapse
|
31
|
Heining E, Bhushan R, Paarmann P, Henis YI, Knaus P. Spatial segregation of BMP/Smad signaling affects osteoblast differentiation in C2C12 cells. PLoS One 2011; 6:e25163. [PMID: 21998639 PMCID: PMC3187766 DOI: 10.1371/journal.pone.0025163] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Accepted: 08/26/2011] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Bone morphogenetic proteins (BMPs) are involved in a plethora of cellular processes in embryonic development and adult tissue homeostasis. Signaling specificity is achieved by dynamic processes involving BMP receptor oligomerization and endocytosis. This allows for spatiotemporal control of Smad dependent and non-Smad pathways. In this study, we investigate the spatiotemporal regulation within the BMP-induced Smad transcriptional pathway. METHODOLOGY/PRINCIPAL FINDINGS Here we discriminate between Smad signaling events that are dynamin-dependent (i.e., require an intact endocytic pathway) and dynamin-independent. Inhibition of dynamin-dependent endocytosis in fluorescence microscopy and fractionation studies revealed a delay in Smad1/5/8 phosphorylation and nuclear translocation after BMP-2 stimulation of C2C12 cells. Using whole genome microarray and qPCR analysis, we identified two classes of BMP-2 induced genes that are differentially affected by inhibition of endocytosis. Thus, BMP-2 induced gene expression of Id1, Id3, Dlx2 and Hey1 is endocytosis-dependent, whereas BMP-2 induced expression of Id2, Dlx3, Zbtb2 and Krt16 is endocytosis-independent. Furthermore, we demonstrate that short term inhibition of endocytosis interferes with osteoblast differentiation as measured by alkaline phosphatase (ALP) production and qPCR analysis of osteoblast marker gene expression. CONCLUSIONS/SIGNIFICANCE Our study demonstrates that dynamin-dependent endocytosis is crucial for the concise spatial activation of the BMP-2 induced signaling cascade. Inhibition of endocytic processes during BMP-2 stimulation leads to altered Smad1/5/8 signaling kinetics and results in differential target gene expression. We show that interfering with the BMP-2 induced transcriptional network by endocytosis inhibition results in an attenuation of osteoblast differentiation. This implies that selective sensitivity of gene expression to endocytosis provides an additional mechanism for the cell to respond to BMP in a context specific manner. Moreover, we suggest a novel Smad dependent signal cascade induced by BMP-2, which does not require endocytosis.
Collapse
Affiliation(s)
- Eva Heining
- Institute for Chemistry and Biochemistry, Freie Universitaet Berlin, Berlin, Germany
| | - Raghu Bhushan
- Institute for Chemistry and Biochemistry, Freie Universitaet Berlin, Berlin, Germany
| | - Pia Paarmann
- Institute for Chemistry and Biochemistry, Freie Universitaet Berlin, Berlin, Germany
| | - Yoav I. Henis
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universitaet Berlin, Berlin, Germany
- * E-mail:
| |
Collapse
|
32
|
Motyl KJ, Rosen CJ. Temperatures rising: brown fat and bone. DISCOVERY MEDICINE 2011; 11:179-185. [PMID: 21447277 PMCID: PMC3629549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Caloric restriction is associated with a reduction in body weight and temperature, as well as a reduction in trabecular bone volume and paradoxically an increase in adipocytes within the bone marrow. The nature of these adipocytes is uncertain, although there is emerging evidence of a direct relationship between bone remodeling and brown adipocytes. For example, in heterotrophic ossification, brown adipocytes set up a hypoxic gradient that leads to vascular invasion, chondrocyte differentiation, and subsequent bone formation. Additionally, deletion of retinoblastoma protein in an osteosarcoma model leads to increased hibernoma (brown fat tumor). Brown adipose tissue (BAT) becomes senescent with age at a time when thermoregulation is altered, bone loss becomes apparent, and sympathetic activity increases. Interestingly, heart rate is an unexpected but good predictor of fracture risk in elderly individuals, pointing to a key role for the sympathetic nervous system in senile osteoporosis. Hence the possibility exists that BAT could play an indirect role in age-related bone loss. However, evidence of an indirect effect from thermogenic dysfunction on bone loss is currently limited. Here, we present current evidence for a relationship between brown adipose tissue and bone as well as provide novel insights into the effects of thermoregulation on bone mineral density.
Collapse
Affiliation(s)
- Katherine J. Motyl
- Maine Medical Center Research Institute 81 Research Drive Scarborough, ME 04074
| | - Clifford J. Rosen
- Maine Medical Center Research Institute 81 Research Drive Scarborough, ME 04074
| |
Collapse
|
33
|
Motyl KJ, Raetz M, Tekalur SA, Schwartz RC, McCabe LR. CCAAT/enhancer binding protein β-deficiency enhances type 1 diabetic bone phenotype by increasing marrow adiposity and bone resorption. Am J Physiol Regul Integr Comp Physiol 2011; 300:R1250-60. [PMID: 21346244 DOI: 10.1152/ajpregu.00764.2010] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bone loss in type 1 diabetes is accompanied by increased marrow fat, which could directly reduce osteoblast activity or result from altered bone marrow mesenchymal cell lineage selection (adipocyte vs. osteoblast). CCAAT/enhancer binding protein beta (C/EBPβ) is an important regulator of both adipocyte and osteoblast differentiation. C/EBPβ-null mice have delayed bone formation and defective lipid accumulation in brown adipose tissue. To examine the balance of C/EBPβ functions in the diabetic context, we induced type 1 diabetes in C/EBPβ-null (knockout, KO) mice. We found that C/EBPβ deficiency actually enhanced the diabetic bone phenotype. While KO mice had reduced peripheral fat mass compared with wild-type mice, they had 5-fold more marrow adipocytes than diabetic wild-type mice. The enhanced marrow adiposity may be attributed to compensation by C/EBPδ, peroxisome proliferator-activated receptor-γ2, and C/EBPα. Concurrently, we observed reduced bone density. Relative to genotype controls, trabecular bone volume fraction loss was escalated in diabetic KO mice (-48%) compared with changes in diabetic wild-type mice (-22%). Despite greater bone loss, osteoblast markers were not further suppressed in diabetic KO mice. Instead, osteoclast markers were increased in the KO diabetic mice. Thus, C/EBPβ deficiency increases diabetes-induced bone marrow (not peripheral) adipose depot mass, and promotes additional bone loss through stimulating bone resorption. C/EBPβ-deficiency also reduced bone stiffness and diabetes exacerbated this (two-way ANOVA P < 0.02). We conclude that C/EBPβ alone is not responsible for the bone vs. fat phenotype switch observed in T1 diabetes and that suppression of CEBPβ levels may further bone loss and decrease bone stiffness by increasing bone resorption.
Collapse
Affiliation(s)
- Katherine J Motyl
- Department of Biomedical and Integrative Physiology, Biomedical Imaging Research Center, Michigan State University, East Lansing, MI 48824, USA
| | | | | | | | | |
Collapse
|
34
|
Rapamycin and the transcription factor C/EBPbeta as a switch in osteoclast differentiation: implications for lytic bone diseases. J Mol Med (Berl) 2009; 88:227-33. [PMID: 19943027 PMCID: PMC2836244 DOI: 10.1007/s00109-009-0567-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2009] [Revised: 10/23/2009] [Accepted: 11/02/2009] [Indexed: 10/24/2022]
Abstract
Lytic bone diseases and in particular osteoporosis are common age-related diseases characterized by enhanced bone fragility due to loss of bone density. Increasingly, osteoporosis poses a major global health-care problem due to the growth of the elderly population. Recently, it was found that the gene regulatory transcription factor CCAAT/enhancer binding protein beta (C/EBPbeta) is involved in bone metabolism. C/EBPbeta occurs as different protein isoforms of variable amino terminal length, and regulation of the C/EBPbeta isoform ratio balance was found to represent an important factor in osteoclast differentiation and bone homeostasis. Interestingly, adjustment of the C/EBPbeta isoform ratio by the process of translational control is downstream of the mammalian target of rapamycin kinase (mTOR), a sensor of the nutritional status and a target of immunosuppressive and anticancer drugs. The findings imply that modulating the process of translational control of C/EBPbeta isoform expression could represent a novel therapeutic approach in osteolytic bone diseases, including cancer and infection-induced bone loss.
Collapse
|
35
|
Transcription factor C/EBPbeta isoform ratio regulates osteoclastogenesis through MafB. EMBO J 2009; 28:1769-81. [PMID: 19440205 PMCID: PMC2685610 DOI: 10.1038/emboj.2009.127] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2008] [Accepted: 04/03/2009] [Indexed: 01/06/2023] Open
Abstract
Disequilibrium between bone-forming osteoblasts and bone-resorbing osteoclasts is central to many bone diseases. Here, we show that dysregulated expression of translationally controlled isoforms of CCAAT/enhancer-binding protein β (C/EBPβ) differentially affect bone mass. Alternative translation initiation that is controlled by the mammalian target of rapamycin (mTOR) pathway generates long transactivating (LAP*, LAP) and a short repressive (LIP) isoforms from a single C/EBPβ transcript. Rapamycin, an inhibitor of mTOR signalling increases the ratio of LAP over LIP and inhibits osteoclastogenesis in wild type (WT) but not in C/EBPβ null (c/ebpβ−/−) or in LIP knock-in (L/L) osteoclast precursors. C/EBPβ mutant mouse strains exhibit increased bone resorption and attenuated expression of MafB, a negative regulator of osteoclastogenesis. Ectopic expression of LAP and LIP in monocytes differentially affect the MafB promoter activity, MafB gene expression and dramatically affect osteoclastogenesis. These data show that mTOR regulates osteoclast formation by modulating the C/EBPβ isoform ratio, which in turn affects osteoclastogenesis by regulating MafB expression.
Collapse
|