101
|
Gustafson B, Hedjazifar S, Gogg S, Hammarstedt A, Smith U. Insulin resistance and impaired adipogenesis. Trends Endocrinol Metab 2015; 26:193-200. [PMID: 25703677 DOI: 10.1016/j.tem.2015.01.006] [Citation(s) in RCA: 254] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 01/19/2015] [Accepted: 01/21/2015] [Indexed: 12/20/2022]
Abstract
The adipose tissue is crucial in regulating insulin sensitivity and risk for diabetes through its lipid storage capacity and thermogenic and endocrine functions. Subcutaneous adipose tissue (SAT) stores excess lipids through expansion of adipocytes (hypertrophic obesity) and/or recruitment of new precursor cells (hyperplastic obesity). Hypertrophic obesity in humans, a characteristic of genetic predisposition for diabetes, is associated with abdominal obesity, ectopic fat accumulation, and the metabolic syndrome (MS), while the ability to recruit new adipocytes prevents this. We review the regulation of adipogenesis, its relation to SAT expandability and the risks of ectopic fat accumulation, and insulin resistance. The actions of GLUT4 in SAT, including a novel family of lipids enhancing insulin sensitivity/secretion, and the function of bone morphogenetic proteins (BMPs) in white and beige/brown adipogenesis in humans are highlighted.
Collapse
Affiliation(s)
- Birgit Gustafson
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy at the University of Gothenburg, SE-41345 Gothenburg, Sweden
| | - Shahram Hedjazifar
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy at the University of Gothenburg, SE-41345 Gothenburg, Sweden
| | - Silvia Gogg
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy at the University of Gothenburg, SE-41345 Gothenburg, Sweden
| | - Ann Hammarstedt
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy at the University of Gothenburg, SE-41345 Gothenburg, Sweden
| | - Ulf Smith
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy at the University of Gothenburg, SE-41345 Gothenburg, Sweden.
| |
Collapse
|
102
|
Qian S, Huang H, Tang Q. Brown and beige fat: the metabolic function, induction, and therapeutic potential. Front Med 2015; 9:162-72. [DOI: 10.1007/s11684-015-0382-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 10/16/2014] [Indexed: 12/27/2022]
|
103
|
Differential lncRNA expression profiles in brown and white adipose tissues. Mol Genet Genomics 2014; 290:699-707. [DOI: 10.1007/s00438-014-0954-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 11/10/2014] [Indexed: 02/03/2023]
|
104
|
Domingues-Faria C, Chanet A, Salles J, Berry A, Giraudet C, Patrac V, Denis P, Bouton K, Goncalves-Mendes N, Vasson MP, Boirie Y, Walrand S. Vitamin D deficiency down-regulates Notch pathway contributing to skeletal muscle atrophy in old wistar rats. Nutr Metab (Lond) 2014; 11:47. [PMID: 25317198 PMCID: PMC4195890 DOI: 10.1186/1743-7075-11-47] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 09/24/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The diminished ability of aged muscle to self-repair is a factor behind sarcopenia and contributes to muscle atrophy. Muscle repair depends on satellite cells whose pool size is diminished with aging. A reduction in Notch pathway activity may explain the age-related decrease in satellite cell proliferation, as this pathway has been implicated in satellite cell self-renewal. Skeletal muscle is a target of vitamin D which modulates muscle cell proliferation and differentiation in vitro and stimulates muscle regeneration in vivo. Vitamin D status is positively correlated to muscle strength/function, and elderly populations develop a vitamin D deficiency. The aim of this study was to evaluate how vitamin D deficiency induces skeletal muscle atrophy in old rats through a reduction in Notch pathway activity and proliferation potential in muscle. METHODS 15-month-old male rats were vitamin D-depleted or not (control) for 9 months (n = 10 per group). Rats were 24-month-old at the end of the experiment. Gene and/or protein expression of markers of proliferation, or modulating proliferation, and of Notch signalling pathway were studied in the tibialis anterior muscle by qPCR and western blot. An unpaired student's t-test was performed to test the effect of the experimental conditions. RESULTS Vitamin D depletion led to a drop in concentrations of plasma 25-hydroxyvitamin D in depleted rats compared to controls (-74%, p < 0.01). Tibialis anterior weight was decreased in D-depleted rats (-25%, p < 0.05). The D-depleted group showed -39%, -31% drops in expression of two markers known to modulate proliferation (Bmp4, Fgf-2 mRNA levels) and -56% drop in one marker of cell proliferation (PCNA protein expression) compared to controls (p < 0.05). Notch pathway activity was blunted in tibialis anterior of D-depleted rats compared to controls, seen as a down-regulation of cleaved Notch (-53%, p < 0.05) and its target Hes1 (-35%, p < 0.05). CONCLUSIONS A 9-month vitamin D depletion induced vitamin D deficiency in old rats. Vitamin D depletion induces skeletal muscle atrophy in old rats through a reduction in Notch pathway activity and proliferation potential. Vitamin D deficiency could aggravate the age-related decrease in muscle regeneration capacity.
Collapse
Affiliation(s)
- Carla Domingues-Faria
- Université d'Auvergne, Unité de Nutrition Humaine, Equipe ECREIN, CLARA, CRNH Auvergne; INRA, UMR 1019, UNH, CRNH Auvergne, Clermont Université, 63000 Clermont-Ferrand, France ; Université d'Auvergne, Unité de Nutrition Humaine, Equipe NuTriM, CRNH Auvergne; INRA, UMR 1019, UNH, CRNH Auvergne, Clermont Université, 63000 Clermont-Ferrand, France ; INRA, UMR1019, UNH, CRNH Auvergne, 63000 Clermont-Ferrand, France
| | - Audrey Chanet
- Université d'Auvergne, Unité de Nutrition Humaine, Equipe NuTriM, CRNH Auvergne; INRA, UMR 1019, UNH, CRNH Auvergne, Clermont Université, 63000 Clermont-Ferrand, France ; INRA, UMR1019, UNH, CRNH Auvergne, 63000 Clermont-Ferrand, France
| | - Jérôme Salles
- Université d'Auvergne, Unité de Nutrition Humaine, Equipe NuTriM, CRNH Auvergne; INRA, UMR 1019, UNH, CRNH Auvergne, Clermont Université, 63000 Clermont-Ferrand, France ; INRA, UMR1019, UNH, CRNH Auvergne, 63000 Clermont-Ferrand, France
| | - Alexandre Berry
- Université d'Auvergne, Unité de Nutrition Humaine, Equipe NuTriM, CRNH Auvergne; INRA, UMR 1019, UNH, CRNH Auvergne, Clermont Université, 63000 Clermont-Ferrand, France ; INRA, UMR1019, UNH, CRNH Auvergne, 63000 Clermont-Ferrand, France
| | - Christophe Giraudet
- Université d'Auvergne, Unité de Nutrition Humaine, Equipe NuTriM, CRNH Auvergne; INRA, UMR 1019, UNH, CRNH Auvergne, Clermont Université, 63000 Clermont-Ferrand, France ; INRA, UMR1019, UNH, CRNH Auvergne, 63000 Clermont-Ferrand, France
| | - Véronique Patrac
- Université d'Auvergne, Unité de Nutrition Humaine, Equipe NuTriM, CRNH Auvergne; INRA, UMR 1019, UNH, CRNH Auvergne, Clermont Université, 63000 Clermont-Ferrand, France ; INRA, UMR1019, UNH, CRNH Auvergne, 63000 Clermont-Ferrand, France
| | - Philippe Denis
- Université d'Auvergne, Unité de Nutrition Humaine, Installation Expérimentale de Nutrition, CRNH Auvergne; INRA, UMR 1019, UNH, CRNH Auvergne, Clermont Université, 63000 Clermont-Ferrand, France ; INRA, UMR1019, UNH, CRNH Auvergne, 63000 Clermont-Ferrand, France
| | - Katia Bouton
- Université d'Auvergne, Unité de Nutrition Humaine, Equipe NuTriM, CRNH Auvergne; INRA, UMR 1019, UNH, CRNH Auvergne, Clermont Université, 63000 Clermont-Ferrand, France ; INRA, UMR1019, UNH, CRNH Auvergne, 63000 Clermont-Ferrand, France
| | - Nicolas Goncalves-Mendes
- Université d'Auvergne, Unité de Nutrition Humaine, Equipe ECREIN, CLARA, CRNH Auvergne; INRA, UMR 1019, UNH, CRNH Auvergne, Clermont Université, 63000 Clermont-Ferrand, France
| | - Marie-Paule Vasson
- Université d'Auvergne, Unité de Nutrition Humaine, Equipe ECREIN, CLARA, CRNH Auvergne; INRA, UMR 1019, UNH, CRNH Auvergne, Clermont Université, 63000 Clermont-Ferrand, France ; Centre Jean Perrin, Unité de Nutrition, 63000 Clermont-Ferrand, France
| | - Yves Boirie
- Université d'Auvergne, Unité de Nutrition Humaine, Equipe NuTriM, CRNH Auvergne; INRA, UMR 1019, UNH, CRNH Auvergne, Clermont Université, 63000 Clermont-Ferrand, France ; CHU Clermont-Ferrand, Service de Nutrition Clinique, 63003 Clermont-Ferrand, France
| | - Stéphane Walrand
- Université d'Auvergne, Unité de Nutrition Humaine, Equipe NuTriM, CRNH Auvergne; INRA, UMR 1019, UNH, CRNH Auvergne, Clermont Université, 63000 Clermont-Ferrand, France ; INRA, UMR1019, UNH, CRNH Auvergne, 63000 Clermont-Ferrand, France
| |
Collapse
|
105
|
Carreira AC, Alves GG, Zambuzzi WF, Sogayar MC, Granjeiro JM. Bone Morphogenetic Proteins: structure, biological function and therapeutic applications. Arch Biochem Biophys 2014; 561:64-73. [PMID: 25043976 DOI: 10.1016/j.abb.2014.07.011] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 07/01/2014] [Accepted: 07/08/2014] [Indexed: 01/09/2023]
Abstract
Bone Morphogenetic Proteins (BMPs) are multifunctional secreted cytokines, which belong to the TGF-β superfamily. These glycoproteins act as a disulfide-linked homo- or heterodimers, being potent regulators of bone and cartilage formation and repair, cell proliferation during embryonic development and bone homeostasis in the adult. BMPs are promising molecules for tissue engineering and bone therapy. The present review discusses this family of proteins, their structure and biological function, their therapeutic applications and drawbacks, their effects on mesenchymal stem cells differentiation, and the cell signaling pathways involved in this process.
Collapse
Affiliation(s)
- Ana Claudia Carreira
- Chemistry Institute, Biochemistry Department, University of São Paulo, São Paulo 05508-000, Brazil; NUCEL-NETCEM Cell and Molecular Therapy Center, Medical Clinics Department, School of Medicine, University of São Paulo, São Paulo, 05508-000 SP, Brazil.
| | - Gutemberg Gomes Alves
- Cell and Molecular Biology Department, Institute of Biology, Fluminense Federal University, Niterói, RJ, Brazil.
| | - William Fernando Zambuzzi
- Department of Chemistry and Biochemistry, Biosciences Institute, UNESP: Universidade Estadual Paulista, Botucatu, SP, Brazil.
| | - Mari Cleide Sogayar
- Chemistry Institute, Biochemistry Department, University of São Paulo, São Paulo 05508-000, Brazil; NUCEL-NETCEM Cell and Molecular Therapy Center, Medical Clinics Department, School of Medicine, University of São Paulo, São Paulo, 05508-000 SP, Brazil.
| | - José Mauro Granjeiro
- Bioengineering Division, National Institute of Metrology, Quality, and Technology, Duque de Caxias, RJ, Brazil; Department of Dental Materials, Dental School, Fluminense Federal University, Niteroi, RJ, Brazil.
| |
Collapse
|
106
|
Saben J, Thakali KM, Lindsey FE, Zhong Y, Badger TM, Andres A, Shankar K. Distinct adipogenic differentiation phenotypes of human umbilical cord mesenchymal cells dependent on adipogenic conditions. Exp Biol Med (Maywood) 2014; 239:1340-51. [PMID: 24951473 DOI: 10.1177/1535370214539225] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The umbilical cord (UC) matrix is a source of multipotent mesenchymal stem cells (MSCs) that have adipogenic potential and thus can be a model to study adipogenesis. However, existing variability in adipocytic differentiation outcomes may be due to discrepancies in methods utilized for adipogenic differentiation. Additionally, functional characterization of UCMSCs as adipocytes has not been described. We tested the potential of three well-established adipogenic cocktails containing IBMX, dexamethasone, and insulin (MDI) plus indomethacin (MDI-I) or rosiglitazone (MDI-R) to stimulate adipocyte differentiation in UCMSCs. MDI, MDI-I, and MDI-R treatment significantly increased peroxisome proliferator-activated receptor gamma (PPARγ) and CCAAT-enhancer binding protein alpha (C/EBPα) mRNA and induced lipid droplet formation. However, MDI-I had the greatest impact on mRNA expression of PPARγ, C/EBPα, FABP4, GPD1, PLIN1, PLIN2, and ADIPOQ and lipid accumulation, whereas MDI showed the least. Interestingly, there were no treatment group differences in the amount of PPARγ protein. However, MDI-I treated cells had significantly more C/EBPα protein compared to MDI or MDI-R, suggesting that indomethacin-dependent increased C/EBPα may contribute to the adipogenesis-inducing potency of MDI-I. Additionally, bone morphogenetic protein 4 (BMP4) treatment of UCMSCs did not enhance responsiveness to MDI-induced differentiation. Finally to characterize adipocyte function, differentiated UCMSCs were stimulated with insulin and downstream signaling was assessed. Differentiated UCMSCs were responsive to insulin at two weeks but showed decreased sensitivity by five weeks following differentiation, suggesting that long-term differentiation may induce insulin resistance. Together, these data indicate that UCMSCs undergo adipogenesis when differentiated in MDI, MDI-I, and MDI-R, however the presence of indomethacin greatly enhances their adipogenic potential beyond that of rosiglitazone. Furthermore, our results suggest that insulin signaling pathways of differentiated UCMSCs are functionally similar to adipocytes.
Collapse
Affiliation(s)
- Jessica Saben
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Keshari M Thakali
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Forrest E Lindsey
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Ying Zhong
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Thomas M Badger
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Aline Andres
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Kartik Shankar
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| |
Collapse
|
107
|
Abstract
Adipocyte differentiation, termed adipogenesis, is a complicated process in which pluripotent mesenchymal stem cells differentiate into mature adipocytes. The process of adipocyte differentiation is tightly regulated by a number of transcription factors, hormones and signaling pathway molecules. Recent studies have demonstrated that microRNAs, which belong to small noncoding RNA species, are also involved in adipocyte differentiation. In vivo and in vitro studies have revealed that various microRNAs affect adipogenesis by targeting several adipogenic transcription factors and key signaling molecules. In this review, we will summarize the roles of microRNAs in adipogenesis and their target genes associated with each stage of adipocyte differentiation.
Collapse
Affiliation(s)
- You Hwa Son
- Seoul National University School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Sojeong Ka
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Korea
| | - A Young Kim
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Korea
| | - Jae Bum Kim
- Seoul National University School of Biological Sciences, Seoul National University, Seoul, Korea
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Korea
| |
Collapse
|
108
|
Elsen M, Raschke S, Tennagels N, Schwahn U, Jelenik T, Roden M, Romacho T, Eckel J. BMP4 and BMP7 induce the white-to-brown transition of primary human adipose stem cells. Am J Physiol Cell Physiol 2014; 306:C431-40. [DOI: 10.1152/ajpcell.00290.2013] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
While white adipose tissue (AT) is an energy storage depot, brown AT is specialized in energy dissipation. Uncoupling protein 1 (UCP1)-expressing adipocytes with a different origin than classical brown adipocytes have been found in white AT. These “brite” (brown-in-white) adipocytes may represent a therapeutic target to counteract obesity. Bone morphogenetic proteins (BMPs) play a role in the regulation of adipogenesis. Based on studies with murine cells, BMP4 is assumed to induce stem cell commitment to the white adipocyte lineage, whereas BMP7 promotes brown adipogenesis. There is evidence for discrepancies between mouse and human AT. Therefore, we compared the effect of BMP4 and BMP7 on white-to-brown transition in primary human adipose stem cells (hASCs) from subcutaneous AT. Long-term exposure of hASCs to recombinant BMP4 or BMP7 during differentiation increased adipogenesis, as determined by lipid accumulation and peroxisome proliferator-activated receptor-γ (PPARγ) expression. Not only BMP7, but also BMP4, increased UCP1 expression in hASCs and decreased expression of the white-specific marker TCF21. The ability of hASCs to induce UCP1 in response to BMP4 and BMP7 markedly differed between donors and could be related to the expression of the brite marker CD137. However, mitochondrial content and oxygen consumption were not increased in hASCs challenged with BMP4 and BMP7. In conclusion, we showed for the first time that BMP4 has similar effects on white-to-brown transition as BMP7 in our human cell model. Thus the roles of BMP4 and BMP7 in adipogenesis cannot always be extrapolated from murine to human cell models.
Collapse
Affiliation(s)
- Manuela Elsen
- Paul-Langerhans-Group for Integrative Physiology, German Diabetes Center, Düsseldorf, Germany
| | - Silja Raschke
- Paul-Langerhans-Group for Integrative Physiology, German Diabetes Center, Düsseldorf, Germany
| | - Norbert Tennagels
- R&D Diabetes Division, Sanofi-Aventis Deutschland, Frankfurt, Germany
| | - Uwe Schwahn
- R&D Diabetes Division, Sanofi-Aventis Deutschland, Frankfurt, Germany
| | - Tomas Jelenik
- Institute for Clinical Diabetology, German Diabetes Center, Düsseldorf, Germany; and
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Düsseldorf, Germany; and
- Department of Endocrinology and Diabetology, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Tania Romacho
- Paul-Langerhans-Group for Integrative Physiology, German Diabetes Center, Düsseldorf, Germany
| | - Jürgen Eckel
- Paul-Langerhans-Group for Integrative Physiology, German Diabetes Center, Düsseldorf, Germany
| |
Collapse
|
109
|
Xue R, Wan Y, Zhang S, Zhang Q, Ye H, Li Y. Role of bone morphogenetic protein 4 in the differentiation of brown fat-like adipocytes. Am J Physiol Endocrinol Metab 2014; 306:E363-72. [PMID: 24347060 DOI: 10.1152/ajpendo.00119.2013] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
There are two different types of fat present in mammals: white adipose tissue, the primary site of energy storage, and brown adipose tissue, which is specializes in energy expenditure. Factors that specify the developmental fate and function of brown fat are poorly understood. Bone morphogenic proteins (BMPs) play an important role in adipogenesis. While BMP4 is capable of triggering commitment of stem cells to the white adipocyte lineage, BMP7 triggers commitment of progenitor cells to a brown adipocyte lineage and activates brown adipogenesis. To investigate the differential effects of BMPs on the development of adipocytes, C3H10T1/2 pluripotent cells were pretreated with BMP4 and BMP7, followed by different adipogenic induction cocktails. Both BMP4 and BMP7 unexpectedly activated a full program of brown adipogenesis, including induction of the brown fat-defining marker uncoupling protein-1 (UCP1), increasing the expression of early regulators of brown fat fate PRDM16 (PR domain-containing 16) and induction of mitochondrial biogenesis and function. Implantation of BMP4-pretreated C3H10T1/2 cells into nude mice resulted in the development of adipose tissue depots containing UCP1-positive brown adipocytes. Interestingly, BMP4 could also induce brown fat-like adipocytes in both white and brown preadipocytes, thereby decreasing the classical brown adipocyte marker Zic1 and increasing the recently identified beige adipocyte marker TMEM26. The data indicate an important role for BMP4 in promoting brown adipocyte differentiation and thermogenesis in vivo and in vitro and offers a potentially new therapeutic approach for the treatment of obesity.
Collapse
Affiliation(s)
- Ruidan Xue
- Department of Endocrinoloy and Metabolism, Huashan Hospital, Medical College Fudan University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
110
|
Wei X, Li G, Yang X, Ba K, Fu Y, Fu N, Cai X, Li G, Chen Q, Wang M, Lin Y. Effects of bone morphogenetic protein-4 (BMP-4) on adipocyte differentiation from mouse adipose-derived stem cells. Cell Prolif 2014; 46:416-24. [PMID: 23869763 DOI: 10.1111/cpr.12044] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Accepted: 02/17/2013] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES As mesenchymal stem cells (MSCs) can be isolated easily from adipose tissues while retaining their self-renewal and multi-potential differentiation capacities, they hold promising possibilities for being applied extensively in tissue engineering. Bone morphogenetic protein (BMP) family members have been reported to provide instructive signals to MSCs for them to differentiate into several different cell lineages. The study described here aims to investigate whether BMP-4 could promote adipose-derived stem cell (ASC) differentiation into adipocytes under various concentrations. MATERIALS AND METHODS ASCs were isolated from mouse inguinal adipose pads and cultured in vitro. 10 ng/ml and 50 ng/ml BMP-4 were added to adipogenic media for 8 days. Oil red-O staining, reverse transcription/polymerase chain reaction and immunocytofluorescence staining were performed to examine differentiation of the ASCs. RESULTS As indicated by increased expression of adipogenic and lipogenic genes (PPAR-γ, APN and LPL) and proteins, 50 ng/ml BMP-4 seemed to induce mASCs to differentiate into the adipo-lineage compared to 10 ng/ml BMP-4, and control groups. In addition, lipid droplets accumulated within the adipocytes under 50 ng/ml BMP-4 stimulation, as shown by oil red-O staining. CONCLUSIONS Our present study suggests that BMP-4, as an adipo-inducing factor, promoted adipogenesis of ASCs at higher concentrations (50 ng/ml) and can perhaps be considered as a candidate for use in adipose tissue engineering.
Collapse
Affiliation(s)
- X Wei
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
111
|
Lin JC, Tarn WY, Hsieh WK. Emerging role for RNA binding motif protein 4 in the development of brown adipocytes. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:769-79. [PMID: 24389249 DOI: 10.1016/j.bbamcr.2013.12.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2013] [Revised: 12/21/2013] [Accepted: 12/24/2013] [Indexed: 10/25/2022]
Abstract
RNA-binding motif protein 4 (RBM4) reportedly reprograms the tissue-specific splicing network which modulates the development of muscles and pancreatic β-islets. Herein, we report that Rbm4a(-/-) mice exhibited hyperlipidemia accompanied with reduced mass of interscapular brown adipose tissue (iBAT). Elevated RBM4a led to the isoform shift of IR, Ppar-γ, and Pref-1 genes which play pivotal roles in the different stages of adipogenesis. Overexpression of RBM4a enhanced the mitochondrial activity of brown adipocyte-like lineage in the presence of uncoupling agent. RBM4a-ablated adipocytes inversely exhibited impaired development and inefficient energy expenditure. Intriguingly, overexpressed RBM4a induced the expression of brown adipocyte-specific factors (Prdm16 and Bmp7) in white adipocyte-like lineage, which suggested the potential action of RBM4a on the white-to-brown trans-differentiation of adipocytes. In differentiating adipocytes, RBM4a constituted a feed-forward circuit through autoregulating the splicing pattern of its own transcript. Based on these results, we propose the emerging role of RBM4 in regulating the adipocyte-specific splicing events and transcription cascade, which subsequently facilitate the development and function of brown adipocyte-like cells.
Collapse
Affiliation(s)
- Jung-Chun Lin
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
| | - Woan-Yuh Tarn
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Wen-Kou Hsieh
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
112
|
Sun YN, Gao Y, Qiao SP, Wang SZ, Duan K, Wang YX, Li H, Wang N. Epigenetic DNA methylation in the promoters of Peroxisome Proliferator-Activated Receptor γ in chicken lines divergently selected for fatness1. J Anim Sci 2014; 92:48-53. [DOI: 10.2527/jas.2013-6962] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Y. N. Sun
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province; College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, P. R. China
- College of Life Science and Agriculture Forestry, Qiqihar University, Qiqihar, 161006, P. R. China
| | - Y. Gao
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province; College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - S. P. Qiao
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province; College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - S. Z. Wang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province; College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - K. Duan
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province; College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - Y. X. Wang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province; College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - H. Li
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province; College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - N. Wang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province; College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, P. R. China
| |
Collapse
|
113
|
Association of genetic variants of BMP4 with type 2 diabetes mellitus and clinical traits in a Chinese Han population. BIOMED RESEARCH INTERNATIONAL 2013; 2013:238150. [PMID: 24350253 PMCID: PMC3855940 DOI: 10.1155/2013/238150] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 10/16/2013] [Accepted: 10/18/2013] [Indexed: 11/17/2022]
Abstract
BMP4 is one of the transforming growth factor- β superfamily, which can participate in adipogenesis. Gene encoding BMP4 is acknowledged as a convincing candidate that may contribute to both glucose and lipid metabolism. In this paper, we aimed to test the impacts of BMP4 variants on type 2 diabetes in a large sample of Chinese population. We genotyped 10 tagging single nucleotide polymorphisms within the BMP4 region in 6822 participants and acquired detailed clinical investigations and biochemistry measurements. We found that BMP4 rs8014363 showed nominal association towards type 2 diabetes, with the T allele conferring a high risk of type 2 diabetes (OR = 1.108, 95%CI 0.999-1.229, P = 0.051 for allele; OR = 1.110, 95%CI 1.000-1.231, P = 0.050 for genotype), but it was no longer statistically significant after adjusting for multiple testing (empirical P = 0.3689 for allele based on 10,000 permutations). Moreover, we observed a significant association of rs8014363 with triglyceride level and a trend towards association with high-density lipoprotein cholesterol after adjusting for age, gender, and BMI (P = 0.035 and 0.068, resp.). Our data suggested that the genetic variants of BMP4 may not play a dominant role in glucose metabolism in Chinese Han population, but a minor effect cannot be ignored.
Collapse
|
114
|
Birsoy K, Festuccia WT, Laplante M. A comparative perspective on lipid storage in animals. J Cell Sci 2013; 126:1541-52. [PMID: 23658371 DOI: 10.1242/jcs.104992] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Lipid storage is an evolutionary conserved process that exists in all organisms from simple prokaryotes to humans. In Metazoa, long-term lipid accumulation is restricted to specialized cell types, while a dedicated tissue for lipid storage (adipose tissue) exists only in vertebrates. Excessive lipid accumulation is associated with serious health complications including insulin resistance, type 2 diabetes, cardiovascular diseases and cancer. Thus, significant advances have been made over the last decades to dissect out the molecular and cellular mechanisms involved in adipose tissue formation and maintenance. Our current understanding of adipose tissue development comes from in vitro cell culture and mouse models, as well as recent approaches to study lipid storage in genetically tractable lower organisms. This Commentary gives a comparative insight into lipid storage in uni- and multi-cellular organisms with a particular emphasis on vertebrate adipose tissue. We also highlight the molecular mechanisms and nutritional signals that regulate the formation of mammalian adipose tissue.
Collapse
Affiliation(s)
- Kivanç Birsoy
- Whitehead Institute for Biomedical Research, Nine Cambridge Center, Cambridge, MA 02142, USA.
| | | | | |
Collapse
|
115
|
Gustafson B, Hammarstedt A, Hedjazifar S, Smith U. Restricted adipogenesis in hypertrophic obesity: the role of WISP2, WNT, and BMP4. Diabetes 2013; 62:2997-3004. [PMID: 23970518 PMCID: PMC3749334 DOI: 10.2337/db13-0473] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
116
|
Chazenbalk G, Singh P, Irge D, Shah A, Abbott DH, Dumesic DA. Androgens inhibit adipogenesis during human adipose stem cell commitment to preadipocyte formation. Steroids 2013; 78:920-6. [PMID: 23707571 PMCID: PMC3951890 DOI: 10.1016/j.steroids.2013.05.001] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Revised: 03/29/2013] [Accepted: 05/06/2013] [Indexed: 11/28/2022]
Abstract
Androgens play a pivotal role in the regulation of body fat distribution. Adipogenesis is a process whereby multipotent adipose stem cells (ASCs) initially become preadipocytes (ASC commitment to preadipocytes) before differentiating into adipocytes. Androgens inhibit human (h) subcutaneous (SC) abdominal preadipocyte differentiation in both sexes, but their effects on hASC commitment to preadipocyte formation is unknown. We therefore examined whether androgen exposure to human (h) ASCs, isolated from SC abdominal adipose of nonobese women, impairs their commitment to preadipocyte formation and/or subsequent differentiation into adipocytes. For this, isolated hASCs from SC abdominal lipoaspirate were cultured in adipogenesis-inducing medium for 0.5-14days in the presence of testosterone (T, 0-100nM) or dihydrotestosterone (DHT, 0-50nM). Adipogenesis was determined by immunofluorescence microscopy and by quantification of adipogenically relevant transcriptional factors, PPARγ, C/EBPα and C/EBPβ. We found that a 3-day exposure of hASCs to T (50nM) or DHT (5nM) in adipogenesis-inducing medium impaired lipid acquisition and decreased PPARγ, C/EBPα and C/EBPβ gene expression. The inhibitory effects of T and DHT at this early-stage of adipocyte differentiation, were partially and completely reversed by flutamide (F, 100nM), respectively. The effect of androgens on hASC commitment to a preadipocyte phenotype was examined via activation of Bone Morphogenic Protein 4 (BMP4) signaling. T (50nM) and DHT (5nM) significantly inhibited the stimulatory effect of BMP4-induced ASC commitment to the preadipocyte phenotype, as regards PPARγ and C/EBPα gene expression. Our findings indicate that androgens, in part through androgen receptor action, impair BMP4-induced commitment of SC hASCs to preadipocytes and also reduce early-stage adipocyte differentiation, perhaps limiting adipocyte numbers and fat storage in SC abdominal adipose.
Collapse
Affiliation(s)
- Gregorio Chazenbalk
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States.
| | | | | | | | | | | |
Collapse
|
117
|
Suenaga M, Kurosawa N, Asano H, Kanamori Y, Umemoto T, Yoshida H, Murakami M, Kawachi H, Matsui T, Funaba M. Bmp4 expressed in preadipocytes is required for the onset of adipocyte differentiation. Cytokine 2013; 64:138-45. [PMID: 23911203 DOI: 10.1016/j.cyto.2013.07.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2013] [Revised: 07/03/2013] [Accepted: 07/08/2013] [Indexed: 12/19/2022]
Abstract
We previously revealed that endogenous bone morphogenetic protein (Bmp) activity is required for lipid accumulation in 3T3-L1 adipocytes. The present study characterized the role of endogenous Bmp activity in preadipocytes. Endogenous Bmp activity was monitored by analyzing the level of phosphorylation of Smad1/5/8, downstream molecules in the Bmp pathway. Higher levels of phosphorylated Smad1/5/8 were detected in adipogenic cells but not in non-adipogenic cells prior to differentiation induction. The inhibition of the Bmp pathway during this period decreased the expression of Pparγ2 and C/ebpα, which are transcription factors responsible for adipocyte differentiation. The expression of these transcription factors were also down-regulated by Bmp4 knockdown. In addition, endogenous Bmp4 was required for the repression of Intrleukin-11 expression. Endogenous Bmp4 in preadipocytes is indispensable for the onset of the adipogenic program, and may help to maintain the preadipocytic state during adipocyte differentiation.
Collapse
Affiliation(s)
- Masashi Suenaga
- Division of Applied Biosciences, Kyoto University Graduate School of Agriculture, Kyoto 606-8502, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
118
|
Sundelacruz S, Levin M, Kaplan DL. Depolarization alters phenotype, maintains plasticity of predifferentiated mesenchymal stem cells. Tissue Eng Part A 2013; 19:1889-908. [PMID: 23738690 DOI: 10.1089/ten.tea.2012.0425.rev] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Although adult stem cell transplantation has been implemented as a therapy for tissue repair, it is limited by the availability of functional adult stem cells. A potential approach to generate stem and progenitor cells may be to modulate the differentiated status of somatic cells. Therefore, there is a need for a better understanding of how the differentiated phenotype of mature cells is regulated. We hypothesize that bioelectric signaling plays an important role in the maintenance of the differentiated state, as it is a functional regulator of the differentiation process in various cells and tissues. In this study, we asked whether the mature phenotype of osteoblasts and adipocytes derived from human mesenchymal stem cells (hMSCs) could be altered by modulation of their membrane potential. hMSC-derived osteoblasts and adipocytes were depolarized by treatment with ouabain, a Na(+)/K(+) ATPase inhibitor, or by treatment with high concentrations of extracellular K(+). To characterize the effect of voltage modulation on the differentiated state, the depolarized cells were evaluated for (1) the loss of differentiation markers; (2) the up-regulation of stemness markers and stem properties; and (3) differences in gene expression profiles in response to voltage modulation. hMSC-derived osteoblasts and adipocytes exhibited significant down-regulation of bone and fat tissue markers in response to depolarization, despite the presence of differentiation-inducing soluble factors, suggesting that bioelectric signaling overrides biochemical signaling in the maintenance of cell state. Suppression of the osteoblast or adipocyte phenotype was not accompanied by up-regulation of genes associated with the stem state. Thus, depolarization does not activate the stem cell genetic signature and, therefore, does not induce a full reprogramming event. However, after transdifferentiating the depolarized cells to evaluate for multi-lineage potential, depolarized osteoblasts demonstrated improved ability to achieve correct adipocyte morphology compared with nondepolarized osteoblasts. The present study thus demonstrates that depolarization reduces the differentiated phenotype of hMSC-derived cells and improves their transdifferentiation capacity, but does not restore a stem-like genetic profile. Through global transcript profiling of depolarized osteoblasts, we identified pathways that may mediate the effects of voltage signaling on cell state, which will require a detailed mechanistic inquiry in future studies.
Collapse
Affiliation(s)
- Sarah Sundelacruz
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, USA
| | | | | |
Collapse
|
119
|
Lee JS, Ha L, Kwon IK, Lim JY. The role of focal adhesion kinase in BMP4 induction of mesenchymal stem cell adipogenesis. Biochem Biophys Res Commun 2013; 435:696-701. [DOI: 10.1016/j.bbrc.2013.05.045] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 05/10/2013] [Indexed: 10/26/2022]
|
120
|
BMP4-mediated brown fat-like changes in white adipose tissue alter glucose and energy homeostasis. Proc Natl Acad Sci U S A 2013; 110:E798-807. [PMID: 23388637 DOI: 10.1073/pnas.1215236110] [Citation(s) in RCA: 232] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Expression of bone morphogenetic protein 4 (BMP4) in adipocytes of white adipose tissue (WAT) produces "white adipocytes" with characteristics of brown fat and leads to a reduction of adiposity and its metabolic complications. Although BMP4 is known to induce commitment of pluripotent stem cells to the adipocyte lineage by producing cells that possess the characteristics of preadipocytes, its effects on the mature white adipocyte phenotype and function were unknown. Forced expression of a BMP4 transgene in white adipocytes of mice gives rise to reduced WAT mass and white adipocyte size along with an increased number of a white adipocyte cell types with brown adipocyte characteristics comparable to those of beige or brite adipocytes. These changes correlate closely with increased energy expenditure, improved insulin sensitivity, and protection against diet-induced obesity and diabetes. Conversely, BMP4-deficient mice exhibit enlarged white adipocyte morphology and impaired insulin sensitivity. We identify peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC1α) as the target of BMP signaling required for these brown fat-like changes in WAT. This effect of BMP4 on WAT appears to extend to human adipose tissue, because the level of expression of BMP4 in WAT correlates inversely with body mass index. These findings provide a genetic and metabolic basis for BMP4's role in altering insulin sensitivity by affecting WAT development.
Collapse
|
121
|
Modica S, Wolfrum C. Bone morphogenic proteins signaling in adipogenesis and energy homeostasis. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:915-23. [PMID: 23353598 DOI: 10.1016/j.bbalip.2013.01.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2012] [Revised: 01/07/2013] [Accepted: 01/10/2013] [Indexed: 01/07/2023]
Abstract
A great deal is known about the molecular mechanisms regulating terminal differentiation of pre-adipocytes into mature adipocytes. In contrast, the knowledge about pathways that trigger commitment of mesenchymal stem cells into the adipocyte lineage is fragmented. In recent years, the role of members of the bone morphogenic protein family in regulating the early steps of adipogenesis has been the focus of research. Findings based on these studies have also highlighted an unexpected role for some bone morphogenic protein in energy homeostasis via regulation of adipocyte development and function. This review summarizes the knowledge about bone morphogenic proteins and their role in adipocyte commitment and regulation of whole body energy homeostasis. This article is part of a Special Issue entitled Brown and White Fat: From Signaling to Disease.
Collapse
Affiliation(s)
- Salvatore Modica
- Institute of Food, Nutrition and Health, ETH Zurich, Switzerland
| | | |
Collapse
|
122
|
Huang HY, Chen SZ, Zhang WT, Wang SS, Liu Y, Li X, Sun X, Li YM, Wen B, Lei QY, Tang QQ. Induction of EMT-like response by BMP4 via up-regulation of lysyl oxidase is required for adipocyte lineage commitment. Stem Cell Res 2013; 10:278-87. [PMID: 23395997 DOI: 10.1016/j.scr.2012.12.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2012] [Revised: 12/25/2012] [Accepted: 12/26/2012] [Indexed: 01/10/2023] Open
Abstract
The developmental pathway that gives rise to mature adipocytes involves commitment and terminal differentiation. Our previous findings indicate that BMP4 (bone morphogenetic protein 4) induces nearly complete commitment of C3H10T1/2 pluripotent stem cells to the adipocyte lineage and knockdown of lysyl oxidase (Lox) disrupts this commitment process. Here, we found that an epithelial-mesenchymal transition (EMT)-like response is required for adipocyte lineage commitment and that Lox is indispensable for this process. When C3H10T1/2 cells were treated with BMP4, Vim and Cdh2 showed up-regulated expression while Cdh1 and Ocln were down-regulated along with enhanced cell migration, which are EMT-like responses. Silencing of Lox in BMP4-treated C3H10T1/2 cells induced a mesenchymal-epithelial transition (MET)-like response associated with the repression of mesenchymal markers, induction of epithelial markers and decreased cell migration. Importantly, blocking the EMT-like response by knocking down Cdh2 or over-expression of Cdh1 impairs adipocyte lineage commitment. EMT is regulated by distinct transcription factors such as Snai1, Snai2 and Twist. In this study, we also found that only Twist was down-regulated after Lox silencing in C3H10T1/2 cells treated with BMP4. This study provides new insights into adipocyte lineage commitment.
Collapse
Affiliation(s)
- Hai-Yan Huang
- Key Laboratory of Molecular Medicine, Ministry of Education; Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai 200032, PR China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
123
|
James AW. Review of Signaling Pathways Governing MSC Osteogenic and Adipogenic Differentiation. SCIENTIFICA 2013; 2013:684736. [PMID: 24416618 PMCID: PMC3874981 DOI: 10.1155/2013/684736] [Citation(s) in RCA: 309] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 11/21/2013] [Indexed: 05/07/2023]
Abstract
Mesenchymal stem cells (MSC) are multipotent cells, functioning as precursors to a variety of cell types including adipocytes, osteoblasts, and chondrocytes. Between osteogenic and adipogenic lineage commitment and differentiation, a theoretical inverse relationship exists, such that differentiation towards an osteoblast phenotype occurs at the expense of an adipocytic phenotype. This balance is regulated by numerous, intersecting signaling pathways that converge on the regulation of two main transcription factors: peroxisome proliferator-activated receptor- γ (PPAR γ ) and Runt-related transcription factor 2 (Runx2). These two transcription factors, PPAR γ and Runx2, are generally regarded as the master regulators of adipogenesis and osteogenesis. This review will summarize signaling pathways that govern MSC fate towards osteogenic or adipocytic differentiation. A number of signaling pathways follow the inverse balance between osteogenic and adipogenic differentiation and are generally proosteogenic/antiadipogenic stimuli. These include β -catenin dependent Wnt signaling, Hedgehog signaling, and NELL-1 signaling. However, other signaling pathways exhibit more context-dependent effects on adipogenic and osteogenic differentiation. These include bone morphogenic protein (BMP) signaling and insulin growth factor (IGF) signaling, which display both proosteogenic and proadipogenic effects. In summary, understanding those factors that govern osteogenic versus adipogenic MSC differentiation has significant implications in diverse areas of human health, from obesity to osteoporosis to regenerative medicine.
Collapse
Affiliation(s)
- Aaron W. James
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, 10833 Le Conte Avenue, CHS A3-251, Los Angeles, CA 90077, USA
- *Aaron W. James:
| |
Collapse
|
124
|
Fujikura J, Hosoda K, Nakao K. Cell transplantation therapy for diabetes mellitus: endocrine pancreas and adipocyte. Endocr J 2013; 60:697-708. [PMID: 23719783 DOI: 10.1507/endocrj.ej13-0162] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Experimental transplantation of endocrine tissues has led to significant advances in our understanding of endocrinology and metabolism. Endocrine cell transplantation therapy is expected to be applied to the treatment of metabolic endocriopathies. Restoration of functional pancreatic beta-cell mass or of functional adipose mass are reasonable treatment approaches for patients with diabetes or lipodystrophy, respectively. Human induced pluripotent stem (iPS) cell research is having a great impact on life sciences. Doctors Takahashi and Yamanaka discovered that the forced expression of a set of genes can convert mouse and human somatic cells into a pluripotent state [1, 2]. These iPS cells can differentiate into a variety of cell types. Therefore, iPS cells from patients may be a potential cell source for autologous cell replacement therapy. This review briefly summarizes the current knowledge about transplantation therapy for diabetes mellitus, the development of the endocrine pancreas and adipocytes, and endocrine-metabolic disease-specific iPS cells.
Collapse
Affiliation(s)
- Junji Fujikura
- Division of Endocrinology and Metabolism, Kyoto University Hospital, Kyoto 606-8507, Japan.
| | | | | |
Collapse
|
125
|
Melka MG, Gillis J, Bernard M, Abrahamowicz M, Chakravarty MM, Leonard GT, Perron M, Richer L, Veillette S, Banaschewski T, Barker GJ, Büchel C, Conrod P, Flor H, Heinz A, Garavan H, Brühl R, Mann K, Artiges E, Lourdusamy A, Lathrop M, Loth E, Schwartz Y, Frouin V, Rietschel M, Smolka MN, Ströhle A, Gallinat J, Struve M, Lattka E, Waldenberger M, Schumann G, Pavlidis P, Gaudet D, Paus T, Pausova Z. FTO, obesity and the adolescent brain. Hum Mol Genet 2012. [PMID: 23201753 DOI: 10.1093/hmg/dds504] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Genetic variations in fat mass- and obesity (FTO)-associated gene, a well-replicated gene locus of obesity, appear to be associated also with reduced regional brain volumes in elderly. Here, we examined whether FTO is associated with total brain volume in adolescence, thus exploring possible developmental effects of FTO. We studied a population-based sample of 598 adolescents recruited from the French Canadian founder population in whom we measured brain volume by magnetic resonance imaging. Total fat mass was assessed with bioimpedance and body mass index was determined with anthropometry. Genotype-phenotype associations were tested with Merlin under an additive model. We found that the G allele of FTO (rs9930333) was associated with higher total body fat [TBF (P = 0.002) and lower brain volume (P = 0.005)]. The same allele was also associated with higher lean body mass (P = 0.03) and no difference in height (P = 0.99). Principal component analysis identified a shared inverse variance between the brain volume and TBF, which was associated with FTO at P = 5.5 × 10(-6). These results were replicated in two independent samples of 413 and 718 adolescents, and in a meta-analysis of all three samples (n = 1729 adolescents), FTO was associated with this shared inverse variance at P = 1.3 × 10(-9). Co-expression networks analysis supported the possibility that the underlying FTO effects may occur during embryogenesis. In conclusion, FTO is associated with shared inverse variance between body adiposity and brain volume, suggesting that this gene may exert inverse effects on adipose and brain tissues. Given the completion of the overall brain growth in early childhood, these effects may have their origins during early development.
Collapse
Affiliation(s)
- Melkaye G Melka
- The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
126
|
Petrighi Polidori G, Lomax MA, Docherty K. Palmitate enhances the differentiation of mouse embryonic stem cells towards white adipocyte lineages. Mol Cell Endocrinol 2012; 361:40-50. [PMID: 22484460 DOI: 10.1016/j.mce.2012.03.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Revised: 03/02/2012] [Accepted: 03/16/2012] [Indexed: 11/23/2022]
Abstract
The number of adipocyte progenitors is determined early in foetal and neonatal development in a process which may be altered by gender and excess nutrient intake, and which in turn determines fat mass in adulthood and the risk of developing obesity. Here we investigate the hypothesis that excess nutrients, in this case the long chain fatty acid palmitate, can program differentiating stem cells towards white fat lineages. The experiments were performed on mouse embryonic stem cells in chemically defined media (CDM) supplemented with bone morphogenetic protein 4 (BMP4) and all trans-retinoic acid (RA). Subsequent treatment for 21 days with palmitate not only promoted the expression of adipocyte markers and monolocular lipid deposition as observed by RT/QPCR and immunocytochemistry, but also stimulated a considerable enrichment in adipocytes as measured by flow cytometry and a lipolytic response to catecholamines. Palmitate increased protein levels of adiponectin that is preferentially expressed in subcutaneous fat, while inhibiting IGFBP2 and IGFBP3 that are associated with visceral fat. In keeping with this finding, palmitate also increased expression of the subcutaneous markers Shox2 and Twist1 and oestrogenising enzymes. Collectively, these results suggest that palmitate induces differentiation towards subcutaneous fat and that this could occur through its oestrogenising effects on the preadipocyte, suggesting a role for palmitate in programming fat development towards a metabolically favourable profile.
Collapse
Affiliation(s)
- Gioia Petrighi Polidori
- School of Medical Sciences, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen, UK
| | | | | |
Collapse
|
127
|
Park HG, Bak EJ, Woo GH, Kim JM, Quan Z, Kim JM, Yoon HK, Cheon SH, Yoon G, Yoo YJ, Na Y, Cha JH. Licochalcone E has an antidiabetic effect. J Nutr Biochem 2012; 23:759-67. [DOI: 10.1016/j.jnutbio.2011.03.021] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Revised: 03/03/2011] [Accepted: 03/25/2011] [Indexed: 01/14/2023]
|
128
|
Macotela Y, Emanuelli B, Mori MA, Gesta S, Schulz TJ, Tseng YH, Kahn CR. Intrinsic differences in adipocyte precursor cells from different white fat depots. Diabetes 2012; 61:1691-9. [PMID: 22596050 PMCID: PMC3379665 DOI: 10.2337/db11-1753] [Citation(s) in RCA: 226] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Obesity and body fat distribution are important risk factors for the development of type 2 diabetes and metabolic syndrome. Evidence has accumulated that this risk is related to intrinsic differences in behavior of adipocytes in different fat depots. In the current study, we demonstrate that adipocyte precursor cells (APCs) isolated from visceral and subcutaneous white adipose depots of mice have distinct patterns of gene expression, differentiation potential, and response to environmental and genetic influences. APCs derived from subcutaneous fat differentiate well in the presence of classical induction cocktail, whereas those from visceral fat differentiate poorly but can be induced to differentiate by addition of bone morphogenetic protein (BMP)-2 or BMP-4. This difference correlates with major differences in gene expression signature between subcutaneous and visceral APCs. The number of APCs is higher in obesity-prone C57BL/6 mice than obesity-resistant 129 mice, and the number in both depots is increased by up to 270% by exposure of mice to high-fat diet. Thus, APCs from visceral and subcutaneous depots are dynamic populations, which have intrinsic differences in gene expression, differentiation properties, and responses to environmental/genetic factors. Regulation of these populations may provide a new target for the treatment and prevention of obesity and its metabolic complications.
Collapse
|
129
|
Gustafson B, Smith U. The WNT inhibitor Dickkopf 1 and bone morphogenetic protein 4 rescue adipogenesis in hypertrophic obesity in humans. Diabetes 2012; 61:1217-24. [PMID: 22447857 PMCID: PMC3331742 DOI: 10.2337/db11-1419] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Overweight characterized by inappropriate expansion of adipose cells (hypertrophic obesity) is associated with the metabolic syndrome and is caused by an inability to recruit and differentiate new precursor cells. We examined the role of bone morphogenetic protein 4 (BMP4) and WNT activation in the regulation of human adipose cell differentiation. Cluster of differentiation (CD)14(+)/45(+) and CD31(+) cells were first removed before the remaining stromal vascular cells of human subcutaneous biopsy specimens were differentiated with/without different WNT inhibitors and/or BMP4. Inhibition of WNT and induction of Dickkopf 1 (DKK1) were markers of precursor cells undergoing excellent differentiation. The addition of DKK1 inhibited WNT activation and promoted adipogenesis in cells with a low degree of differentiation. The positive effect of DKK1, inhibiting cellular WNT activation by binding to the Kremen/LDL receptor-related protein receptors, was not seen with inhibitors of secreted WNT ligands. BMP4 increased differentiation, and BMP4 in the presence of DKK1 produced an additive effect. There was an apparent cross-talk between differentiation and commitment because BMP4 expression increased in differentiating adipocytes, and the addition of the BMP4 inhibitor, Noggin, reduced precursor cell differentiation. Thus, differentiated human adipose cells can promote adipogenesis via endogenous BMP4 activation, and the impaired adipogenesis in hypertrophic obesity is mainly due to an inability to suppress canonical WNT and to induce DKK1.
Collapse
|
130
|
Musri MM, Gomis R, Párrizas M. A chromatin perspective of adipogenesis. Organogenesis 2012; 6:15-23. [PMID: 20592861 DOI: 10.4161/org.6.1.10226] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2009] [Accepted: 10/01/2009] [Indexed: 12/11/2022] Open
Abstract
The transcriptional cascade governing adipogenesis has been thoroughly examined throughout the years. Transcription factors PPARγ and C/EBPα are universally recognized as the master regulators of adipocyte differentiation and together they direct the establishment of the gene expression pattern of mature adipose cells. However, this familiar landscape has been considerably broadened in recent years by the identification of novel factors that participate in the regulation of adipogenesis, either favoring or inhibiting it, through their effects on chromatin. Epigenetic signals and chromatin-modifying proteins contribute to adipogenesis and, through regulation of the phenotypic maintenance of the mature adipocytes, to the control of metabolism. In this review we intend to summarize the recently described epigenetic events that participate in adipogenesis and their connections with the main factors that constitute the classical transcriptional cascade.
Collapse
Affiliation(s)
- Melina M Musri
- Endocrinology and Nutrition Unit, IDIBAPS, CIBERDEM, Barcelona, Spain
| | | | | |
Collapse
|
131
|
Abstract
Excessive caloric intake without a rise in energy expenditure promotes adipocyte hyperplasia and adiposity. The rise in adipocyte number is triggered by signaling factors that induce conversion of mesenchymal stem cells (MSCs) to preadipocytes that differentiate into adipocytes. MSCs, which are recruited from the vascular stroma of adipose tissue, provide an unlimited supply of adipocyte precursors. Members of the BMP and Wnt families are key mediators of stem cell commitment to produce preadipocytes. Following commitment, exposure of growth-arrested preadipocytes to differentiation inducers [insulin-like growth factor 1 (IGF1), glucocorticoid, and cyclic AMP (cAMP)] triggers DNA replication and reentry into the cell cycle (mitotic clonal expansion). Mitotic clonal expansion involves a transcription factor cascade, followed by the expression of adipocyte genes. Critical to these events are phosphorylations of the transcription factor CCATT enhancer-binding protein β (C/EBPβ) by MAP kinase and GSK3β to produce a conformational change that gives rise to DNA-binding activity. "Activated" C/EBPβ then triggers transcription of peroxisome proliferator-activated receptor-γ (PPARγ) and C/EBPα, which in turn coordinately activate genes whose expression produces the adipocyte phenotype.
Collapse
Affiliation(s)
- Qi Qun Tang
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | |
Collapse
|
132
|
Abstract
Bone morphogenetic protein (BMP) signaling in diseases is the subject of an overwhelming array of studies. BMPs are excellent targets for treatment of various clinical disorders. Several BMPs have already been shown to be clinically beneficial in the treatment of a variety of conditions, including BMP-2 and BMP-7 that have been approved for clinical application in nonunion bone fractures and spinal fusions. With the use of BMPs increasingly accepted in spinal fusion surgeries, other therapeutic approaches targeting BMP signaling are emerging beyond applications to skeletal disorders. These approaches can further utilize next-generation therapeutic tools such as engineered BMPs and ex vivo- conditioned cell therapies. In this review, we focused to provide insights into such clinical potentials of BMPs in metabolic and vascular diseases, and in cancer. [BMB reports 2011; 44(10): 619-634].
Collapse
Affiliation(s)
- Meejung Kim
- Joint Center for Biosciences at Lee Gil Ya Cancer and Diabetes Research Institute, Gachon University of Medicine and Science, IncheonKorea
| | | |
Collapse
|
133
|
Cawthorn WP, Scheller EL, MacDougald OA. Adipose tissue stem cells meet preadipocyte commitment: going back to the future. J Lipid Res 2012; 53:227-46. [PMID: 22140268 PMCID: PMC3269153 DOI: 10.1194/jlr.r021089] [Citation(s) in RCA: 539] [Impact Index Per Article: 44.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
White adipose tissue (WAT) is perhaps the most plastic organ in the body, capable of regeneration following surgical removal and massive expansion or contraction in response to altered energy balance. Research conducted for over 70 years has investigated adipose tissue plasticity on a cellular level, spurred on by the increasing burden that obesity and associated diseases are placing on public health globally. This work has identified committed preadipocytes in the stromal vascular fraction of adipose tissue and led to our current understanding that adipogenesis is important not only for WAT expansion, but also for maintenance of adipocyte numbers under normal metabolic states. At the turn of the millenium, studies investigating preadipocyte differentiation collided with developments in stem cell research, leading to the discovery of multipotent stem cells within WAT. Such adipose tissue-derived stem cells (ASCs) are capable of differentiating into numerous cell types of both mesodermal and nonmesodermal origin, leading to their extensive investigation from a therapeutic and tissue engineering perspective. However, the insights gained through studying ASCs have also contributed to more-recent progress in attempts to better characterize committed preadipocytes in adipose tissue. Thus, ASC research has gone back to its roots, thereby expanding our knowledge of preadipocyte commitment and adipose tissue biology.
Collapse
Affiliation(s)
- William P Cawthorn
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | |
Collapse
|
134
|
Lee JS, Ha L, Park JH, Lim JY. Mechanical stretch suppresses BMP4 induction of stem cell adipogenesis via upregulating ERK but not through downregulating Smad or p38. Biochem Biophys Res Commun 2012; 418:278-83. [PMID: 22266311 DOI: 10.1016/j.bbrc.2012.01.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Accepted: 01/04/2012] [Indexed: 11/28/2022]
Abstract
Bone morphogenetic proteins (BMPs) are also implicated in the commitment of mesenchymal stem cells (MSCs) toward adipocytes. We tested that stretching of cells may downregulate BMP4 induction of MSC adipogenesis. C3H10T1/2 MSCs were pretreated with BMP4 and induced to differentiate to adipocytes using adipogenic hormonal inducers. To test the stretch effect on BMP4 function, cells were exposed to cyclic tensile stretch (10% strain, 0.25Hz, 120min/day) during the BMP4 pretreatment period. BMP4 induced MSC adipocytic commitment. Stretching during the BMP4 exposure could suppress BMP4 induction of MSC adipogenesis, as assessed by downregulated adipogenic transcription factors (PPARγ, C/EBPα, aP2) and decreased lipid accumulation. BMP4 signaled through Smad1/5/8 and p38MAPK, whereas cell stretch did not affect BMP4-induced activation in Smad or p38. On the other hand, cell stretch triggered significant ERK1/2 phosphorylation relative to BMP4 treatment alone cells. Further, stretch suppression of BMP4-induced MSC adipogenesis was significantly deteriorated if cells were stretched with ERK blocked by PD98059. Combined, these suggest that cell stretch suppresses the BMP4 induction of MSC adipogenesis potentially via upregulating ERK but not through the downregulation of Smad or p38. Our data on inhibiting MSC adipogenesis will be of significant interest for obesity and developmental mechanobiology studies.
Collapse
Affiliation(s)
- Jeong Soon Lee
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | | | | | | |
Collapse
|
135
|
Luo X, Hutley LJ, Webster JA, Kim YH, Liu DF, Newell FS, Widberg CH, Bachmann A, Turner N, Schmitz-Peiffer C, Prins JB, Yang GS, Whitehead JP. Identification of BMP and activin membrane-bound inhibitor (BAMBI) as a potent negative regulator of adipogenesis and modulator of autocrine/paracrine adipogenic factors. Diabetes 2012; 61:124-36. [PMID: 22187378 PMCID: PMC3237663 DOI: 10.2337/db11-0998] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Adipose tissue dysfunction underpins the association of obesity with type 2 diabetes. Adipogenesis is required for the maintenance of adipose tissue function. It involves the commitment and subsequent differentiation of preadipocytes and is coordinated by autocrine, paracrine, and endocrine factors. We previously reported that fibroblast growth factor-1 (FGF-1) primes primary human preadipocytes and Simpson Golabi Behmel syndrome (SGBS) preadipocytes and increases adipogenesis through a cascade involving extracellular signal-related kinase 1/2 (ERK1/2). Here, we aimed to use the FGF-1 system to identify novel adipogenic regulators. Expression profiling revealed bone morphogenetic protein (BMP) and activin membrane-bound inhibitor (BAMBI) as a putative FGF-1 effector. BAMBI is a transmembrane protein and modulator of paracrine factors that regulate adipogenesis, including transforming growth factor (TGF) superfamily members (TGF-β and BMP) and Wnt. Functional investigations established BAMBI as a negative regulator of adipogenesis and modulator of the anti- and proadipogenic effects of Wnt3a, TGF-β1, and BMP-4. Further studies showed that BAMBI expression levels are decreased in a mouse model of diet-induced obesity. Collectively, these findings establish BAMBI as a novel, negative regulator of adipogenesis that can act as a nexus to integrate multiple paracrine signals to coordinate adipogenesis. Alterations in BAMBI may play a role in the (patho)physiology of obesity, and manipulation of BAMBI may present a novel therapeutic approach to improve adipose tissue function.
Collapse
Affiliation(s)
- Xiao Luo
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, People’s Republic of China
- Metabolic Program, Mater Medical Research Institute, Mater Health Services, South Brisbane, Queensland, Australia
- Diamantina Institute for Cancer, Immunology and Metabolic Medicine, Princess Alexandra Hospital, University of Queensland, Brisbane, Queensland, Australia
| | - Louise J. Hutley
- Metabolic Program, Mater Medical Research Institute, Mater Health Services, South Brisbane, Queensland, Australia
- Diamantina Institute for Cancer, Immunology and Metabolic Medicine, Princess Alexandra Hospital, University of Queensland, Brisbane, Queensland, Australia
| | - Julie A. Webster
- Metabolic Program, Mater Medical Research Institute, Mater Health Services, South Brisbane, Queensland, Australia
- Diamantina Institute for Cancer, Immunology and Metabolic Medicine, Princess Alexandra Hospital, University of Queensland, Brisbane, Queensland, Australia
| | - Yu-Hee Kim
- Metabolic Program, Mater Medical Research Institute, Mater Health Services, South Brisbane, Queensland, Australia
- Diamantina Institute for Cancer, Immunology and Metabolic Medicine, Princess Alexandra Hospital, University of Queensland, Brisbane, Queensland, Australia
| | - Dong-Fang Liu
- Metabolic Program, Mater Medical Research Institute, Mater Health Services, South Brisbane, Queensland, Australia
- Diamantina Institute for Cancer, Immunology and Metabolic Medicine, Princess Alexandra Hospital, University of Queensland, Brisbane, Queensland, Australia
| | - Felicity S. Newell
- Diamantina Institute for Cancer, Immunology and Metabolic Medicine, Princess Alexandra Hospital, University of Queensland, Brisbane, Queensland, Australia
| | - Charlotte H. Widberg
- Diamantina Institute for Cancer, Immunology and Metabolic Medicine, Princess Alexandra Hospital, University of Queensland, Brisbane, Queensland, Australia
| | - Anthony Bachmann
- Diamantina Institute for Cancer, Immunology and Metabolic Medicine, Princess Alexandra Hospital, University of Queensland, Brisbane, Queensland, Australia
| | - Nigel Turner
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | | | - Johannes B. Prins
- Metabolic Program, Mater Medical Research Institute, Mater Health Services, South Brisbane, Queensland, Australia
- Diamantina Institute for Cancer, Immunology and Metabolic Medicine, Princess Alexandra Hospital, University of Queensland, Brisbane, Queensland, Australia
| | - Gong-She Yang
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, People’s Republic of China
- Corresponding authors: Jonathan P. Whitehead, , and Gong-She Yang,
| | - Jonathan P. Whitehead
- Metabolic Program, Mater Medical Research Institute, Mater Health Services, South Brisbane, Queensland, Australia
- Diamantina Institute for Cancer, Immunology and Metabolic Medicine, Princess Alexandra Hospital, University of Queensland, Brisbane, Queensland, Australia
- Corresponding authors: Jonathan P. Whitehead, , and Gong-She Yang,
| |
Collapse
|
136
|
Markowski DN, Helmke BM, Meyer F, von Ahsen I, Nimzyk R, Nolte I, Bullerdiek J. BMP4 increases expression of HMGA2 in mesenchymal stem cells. Cytokine 2011; 56:811-6. [PMID: 22019704 DOI: 10.1016/j.cyto.2011.09.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Revised: 09/22/2011] [Accepted: 09/25/2011] [Indexed: 10/16/2022]
Abstract
BMP4 has been linked to early steps of adipocyte lineage differentiation but only little is known about its corresponding downstream pathways. Herein, we have investigated whether or not the expression of high mobility group protein HMGA2, another protein linked to proliferation and differentiation within the process of adipogenesis, may be influenced by BMP4 signaling in adipose tissue derived stem cells. Compared to FGF1, a strong inducer of HMGA2 in immortalized pre-adipocytes, BMP4 was found moderately to induce the HMGA2 mRNA expression in serum starved adipose tissue derived stem cells and myometrial cells. In contrast, no such activity was noted in canine bone marrow derived mesenchymal stem cells. As to adipocyte lineage differentiation the functions of BMP4 and HMGA2 mechanistically overlap. Thus, we propose that in adipose tissue BMP4 acts in part by activating HMGA2 making this architectural transcription factor one of the major downstream players in that system.
Collapse
Affiliation(s)
- Dominique Nadine Markowski
- Center of Human Genetics, University of Bremen, Leobener Strasse ZHG, D-28359 Bremen, Germany; Small Animal Clinic, University of Veterinary Medicine and Research Cluster of Excellence "REBIRTH", Bünteweg 9, D-30559 Hannover, Germany
| | | | | | | | | | | | | |
Collapse
|
137
|
Lafontan M. Historical perspectives in fat cell biology: the fat cell as a model for the investigation of hormonal and metabolic pathways. Am J Physiol Cell Physiol 2011; 302:C327-59. [PMID: 21900692 DOI: 10.1152/ajpcell.00168.2011] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
For many years, there was little interest in the biochemistry or physiology of adipose tissue. It is now well recognized that adipocytes play an important dynamic role in metabolic regulation. They are able to sense metabolic states via their ability to perceive a large number of nervous and hormonal signals. They are also able to produce hormones, called adipokines, that affect nutrient intake, metabolism and energy expenditure. The report by Rodbell in 1964 that intact fat cells can be obtained by collagenase digestion of adipose tissue revolutionized studies on the hormonal regulation and metabolism of the fat cell. In the context of the advent of systems biology in the field of cell biology, the present seems an appropriate time to look back at the global contribution of the fat cell to cell biology knowledge. This review focuses on the very early approaches that used the fat cell as a tool to discover and understand various cellular mechanisms. Attention essentially focuses on the early investigations revealing the major contribution of mature fat cells and also fat cells originating from adipose cell lines to the discovery of major events related to hormone action (hormone receptors and transduction pathways involved in hormonal signaling) and mechanisms involved in metabolite processing (hexose uptake and uptake, storage, and efflux of fatty acids). Dormant preadipocytes exist in the stroma-vascular fraction of the adipose tissue of rodents and humans; cell culture systems have proven to be valuable models for the study of the processes involved in the formation of new fat cells. Finally, more recent insights into adipocyte secretion, a completely new role with major metabolic impact, are also briefly summarized.
Collapse
Affiliation(s)
- Max Lafontan
- Institut National de la Santé et de la Recherche Médicale, UMR, Hôpital Rangueil, Toulouse, France.
| |
Collapse
|
138
|
Zamani N, Brown CW. Emerging roles for the transforming growth factor-{beta} superfamily in regulating adiposity and energy expenditure. Endocr Rev 2011; 32:387-403. [PMID: 21173384 PMCID: PMC3365795 DOI: 10.1210/er.2010-0018] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Accepted: 11/23/2010] [Indexed: 12/12/2022]
Abstract
Members of the TGF-β superfamily regulate many aspects of development, including adipogenesis. Studies in cells and animal models have characterized the effects of superfamily signaling on adipocyte development, adiposity, and energy expenditure. Although bone morphogenetic protein (BMP) 4 is generally considered a protein that promotes the differentiation of white adipocytes, BMP7 has emerged as a selective regulator of brown adipogenesis. Conversely, TGF-β and activin A inhibit adipocyte development, a process augmented in TGF-β-treated cells by Smads 6 and 7, negative regulators of canonical TGF-β signaling. Other superfamily members have mixed effects on adipogenesis depending on cell culture conditions, the timing of expression, and the cell type, and many of these effects occur by altering the expression or activities of proteins that control the adipogenic cascade, including members of the CCAAT/enhancer binding protein family and peroxisome proliferator-activated receptor-γ. BMP7, growth differentiation factor (GDF) 8, and GDF3 are versatile in their mechanisms of action, and altering their normal expression characteristics has significant effects on adiposity in vivo. In addition to their roles in adipogenesis, activins and BMP7 regulate energy expenditure by affecting the expression of genes that contribute to mitochondrial biogenesis and function. GDF8 signals through its own receptors during adipogenesis while antagonizing BMP7, an example of a ligand from one major branch of the superfamily regulating the other. With such intricate relationships that ultimately affect adiposity, TGF-β superfamily signaling holds considerable promise as a target for treating human obesity and its comorbidities.
Collapse
Affiliation(s)
- Nader Zamani
- Baylor College of Medicine, Houston, Texas 77030, USA
| | | |
Collapse
|
139
|
Lee JS, Park JH, Kwon IK, Lim JY. Retinoic acid inhibits BMP4-induced C3H10T1/2 stem cell commitment to adipocyte via downregulating Smad/p38MAPK signaling. Biochem Biophys Res Commun 2011; 409:550-5. [PMID: 21605549 DOI: 10.1016/j.bbrc.2011.05.042] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Accepted: 05/06/2011] [Indexed: 12/20/2022]
Abstract
Increased adipocyte formation from mesenchymal stem cells (MSCs) is typical for obesity. It is recently observed that bone morphogenetic proteins (BMPs) provide instructive signals for the commitment of MSCs to adipocytes. We examined potential role of retinoic acid (RA) in inhibiting the BMP4 induction of MSC commitment toward adipocyte. BMP4-treated C3H10T1/2 MSCs, when further exposed to adipogenic differentiation media, displayed distinct adipocytic commitment and differentiation. This could be inhibited by RA exposure during the BMP4 treatment stage (commitment stage before adipogenic hormonal inducers were given), as was observed by reductions in key adipogenic genes/transcription factors (C/EBPα, PPARγ, aP2), lipogenic genes (LPL, FAS, GLUT4), and lipid accumulation. Among RA receptors (RARs) screened, RARβ was mainly upregulated under RA exposure. BMP4 signaled through both Smad1/5/8 and p38 mitogen-activated protein kinase (MAPK) and RA significantly suppressed the BMP4-triggered phosphorylation of both Smad1/5/8 and p38MAPK. These data suggest that RA has inhibitory effects on the BMP4 induction of C3H10T1/2 adipocytic commitment via downregulating Smad/p38MAPK signaling. How to inhibit MSC adipocytic commitment, as partly revealed in this study, will have a significant impact on treating obesity and related diseases.
Collapse
Affiliation(s)
- Jeong Soon Lee
- Department of Engineering Mechanics, College of Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | | | | | | |
Collapse
|
140
|
Defective osteogenic differentiation in the development of osteosarcoma. Sarcoma 2011; 2011:325238. [PMID: 21437219 PMCID: PMC3061279 DOI: 10.1155/2011/325238] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Revised: 12/19/2010] [Accepted: 12/20/2010] [Indexed: 12/21/2022] Open
Abstract
Osteosarcoma (OS) is associated with poor prognosis due to its high incidence of metastasis and chemoresistance. It often arises in areas of rapid bone growth in long bones during the adolescent growth spurt. Although certain genetic conditions and alterations increase the risk of developing OS, the molecular pathogenesis is poorly understood. Recently, defects in differentiation have been linked to cancers, as they are associated with high cell proliferation. Treatments overcoming these defects enable terminal differentiation and subsequent tumor inhibition. OS development may be associated with defects in osteogenic differentiation. While early regulators of osteogenesis are unable to bypass these defects, late osteogenic regulators, including Runx2 and Osterix, are able to overcome some of the defects and inhibit tumor propagation through promoting osteogenic differentiation. Further understanding of the relationship between defects in osteogenic differentiation and tumor development holds tremendous potential in treating OS.
Collapse
|
141
|
Styner M, Sen B, Xie Z, Case N, Rubin J. Indomethacin promotes adipogenesis of mesenchymal stem cells through a cyclooxygenase independent mechanism. J Cell Biochem 2011; 111:1042-50. [PMID: 20672310 DOI: 10.1002/jcb.22793] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Regulation of mesenchymal stem cell (MSC) lineage selection is important for the generation of bone mass. Inhibition of cyclooxygenase-2 (COX2) may increase adipogenesis at the cost of decreasing osteoprogenitor output. Here we investigated the role of COX2 and its products during MSC differentiation. Indomethacin stimulated adipogenesis (increased aP2, adiponectin and lipid droplets) of CH310T1/2 stem cells as well as marrow-derived MSCs to a degree similar to the PPARγ2 ligand, rosiglitazone. Unlike rosiglitazone, indomethacin significantly upregulated PPARγ2 expression. Indomethacin and the COX2 specific inhibitor celecoxib suppressed PGE2 production, but celecoxib did not induce adipogenesis. As well, addition of PGE2 failed to reverse indomethacin induced adipogenesis, indicating that indomethacin's effects were prostaglandin independent. In MSCs over-expressing PPARγ2 and RXRα, indomethacin did not increase PPAR-induced transcription, while rosiglitazone and 15d-PGJ2 did (1.7- and 1.3-fold, respectively, P < 0.001). We considered whether indomethacin might directly affect C/EBPβ proximally to PPARγ2 induction. Indomethacin significantly increased C/EBPβ expression and protein within 24 h of addition. These results indicate that indomethacin promotes adipogenesis by increasing C/EBPβ and PPARγ2 expression in a prostaglandin-independent fashion. This effect of indomethacin is pertinent to potential deleterious effects of this commonly used anti-inflammatory drug on bone remodeling and tissue healing.
Collapse
Affiliation(s)
- Maya Styner
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina 27599-7170, USA.
| | | | | | | | | |
Collapse
|
142
|
Li HX, Xiao L, Wang C, Gao JL, Zhai YG. Review: Epigenetic regulation of adipocyte differentiation and adipogenesis. J Zhejiang Univ Sci B 2011; 11:784-91. [PMID: 20872986 DOI: 10.1631/jzus.b0900401] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
It is generally agreed that adipocytes originate from mesenchymal stem cells in what can be divided into two processes: determination and differentiation. In the past decade, many factors associated with epigenetic signals have been proved to be pivotal for the appropriate timing of adipogenesis progression. A large number of coregulators at critical gene promoters set up specific patterns of DNA methylation, histone acetylation and methylation, and nucleosome rearrangement, that act as an epigenetic code to modulate the correct progress of adipocyte differentiation and adipogenesis during adipogenesis. In this review, we focus on the functions and roles of epigenetic processes in preadipocyte differentiation and adipogenesis.
Collapse
Affiliation(s)
- Hong-xing Li
- Key Laboratory for Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | | | | | | | | |
Collapse
|
143
|
Alfaro MP, Saraswati S, Young PP. Molecular mediators of mesenchymal stem cell biology. VITAMINS AND HORMONES 2011; 87:39-59. [PMID: 22127236 DOI: 10.1016/b978-0-12-386015-6.00023-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mesenchymal stem cells (MSCs) have the ability to self-renew and differentiate into multiple lineages making them an appropriate candidate for stem cell therapy. In spite of achieving considerable success in preclinical models, limited success has been achieved in clinical settings with MSCs. A major impediment that is faced is low survival of MSCs in injured tissues following implantation. In order to enhance the reparative properties of MSCs, it is vital to understand the molecular signals that regulate MSC survival and self-renewal. This review assimilates information that characterizes MSCs and mentions their utilization in myocardial infarction therapy. Additionally, our attempt herein is to gather pertinent published information regarding the role of canonical Wnt and BMP signaling in regulating the potential of MSCs to self-renew, proliferate, differentiate, and survive.
Collapse
Affiliation(s)
- Maria P Alfaro
- Department of Pathology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | | |
Collapse
|
144
|
Son JW, Kim MK, Park YM, Baek KH, Yoo SJ, Song KH, Son HS, Yoon KH, Lee WC, Cha BY, Son HY, Kwon HS. Association of serum bone morphogenetic protein 4 levels with obesity and metabolic syndrome in non-diabetic individuals. Endocr J 2011; 58:39-46. [PMID: 21186333 DOI: 10.1507/endocrj.k10e-248] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Bone morphogenetic protein 4 (BMP-4) is involved in the earliest stages of adipocyte differentiation and is recognized as an adipogenic factor for white adipose tissue. The association of serum BMP-4 levels with anthropometric and metabolic parameters has not been previously studied. We aimed to explore the relationship of serum BMP-4 levels with obesity and metabolic syndrome. Serum BMP-4 levels were measured in 104 non-diabetic individuals from the Chungju Metabolic Disease Cohort Study. Anthropometric measurements and components of metabolic syndrome were assessed in all patients. Serum BMP-4 levels were significantly increased in individuals with obesity or metabolic syndrome. After adjusting for age and gender, serum BMP-4 levels were positively correlated with body mass index, waist circumference (WC), waist-to-hip ratio, fasting plasma insulin, homeostasis model assessment index, and triglycerides and were negatively correlated with high-density lipoprotein (HDL) cholesterol. Among these parameters, WC and HDL cholesterol were found to be independent contributing factors for serum BMP-4 levels. Serum BMP-4 levels were also significantly higher in subjects with positive diagnostic criteria for each component of metabolic syndrome. The area under the receiver operating characteristic curve for BMP-4 was 0.661 (P = 0.022, 95% CI = 0.528 to 0.794) and the cut-off value was 2.84 pg/mL. This is the first demonstration that serum BMP-4 levels are associated with adiposity, insulin resistance, and the presence of metabolic syndrome. Whether BMP-4 may be involved in the pathogenesis of obesity and metabolic syndrome deserves further investigation.
Collapse
Affiliation(s)
- Jang-Won Son
- Division of Endocrinology and Metabolism, Department of Internal Medicine, The Catholic University of Korea, #62 Yeoido-dong, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
145
|
Abstract
PURPOSE OF REVIEW The review highlights recent findings regarding the functions of mitochondria in adipocytes, providing an understanding of their central roles in regulating substrate metabolism, energy expenditure, disposal of reactive oxygen species (ROS), and in the pathophysiology of obesity and insulin resistance, as well as roles in the mechanisms that affect adipogenesis and mature adipocyte function. RECENT FINDINGS Nutrient excess leads to mitochondrial dysfunction, which in turn leads to obesity-related pathologies, in part due to the harmful effects of ROS. The recent recognition of 'ectopic' brown adipose in humans suggests that this tissue may play an underappreciated role in the control of energy expenditure. Transcription factors, PGC-1alpha and PRDM16, which regulate brown adipogenesis, and members of the TGF-beta superfamily that modulate this process may be important new targets for antiobesity drugs. SUMMARY Mitochondria play central roles in ATP production, energy expenditure, and disposal of ROS. Excessive energy substrates lead to mitochondrial dysfunction with consequential effects on lipid and glucose metabolism. Adipocytes help to maintain the appropriate balance between energy storage and expenditure and maintaining this balance requires normal mitochondrial function. Many adipokines, including members of the TGF-beta superfamily, and transcriptional coactivators, PGC-1alpha and PRDM16, are important regulators of this process.
Collapse
Affiliation(s)
- Juan C. Bournat
- Department of Molecular and Human Genetics, Houston, TX 77030, USA
| | - Chester W. Brown
- Department of Molecular and Human Genetics, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital, Houston, TX 77030, USA
| |
Collapse
|
146
|
Mauney J, Olsen BR, Volloch V. Matrix remodeling as stem cell recruitment event: a novel in vitro model for homing of human bone marrow stromal cells to the site of injury shows crucial role of extracellular collagen matrix. Matrix Biol 2010; 29:657-63. [PMID: 20828613 PMCID: PMC6817338 DOI: 10.1016/j.matbio.2010.08.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2010] [Revised: 06/28/2010] [Accepted: 08/17/2010] [Indexed: 01/16/2023]
Abstract
The goal of the present study was to devise an in vitro model suitable for investigations of the homing of mesenchymal stem cells to sites of injury. Such a model was designed on the basis of a "transwell" assay, with an insert seeded with human bone marrow stromal cells and a well with a desired cell type. To mimic physiological environment and to simulate "injury", cells in a well were maintained not only on tissue culture plastic but also on collagens I and IV, major matrix components in musculoskeletal and adipose tissues respectively, and subjected to a severe thermal stress. The results obtained showed a massive translocation of bone marrow stromal cells through the inserts' membrane toward the "injury" site. Unexpectedly, it emerged that collagen matrix is essential in producing such a migration. The results obtained suggest that upon injury cells secrete a substance which interacts with collagen matrix to produce a homing agent. The substance in question appears to be a protease and its interaction with the collagen matrix appears to be a digestion of the latter into fragments shown to be chemotactic. Both AEBSF, an inhibitor of serine proteases, and leupeptin, an inhibitor of cysteine proteases as well as of trypsin-like serine proteases, but not the broad spectrum MMP inhibitor marimastat, significantly inhibit the observed homing effect and this inhibition is not due to cytotoxicity. Moreover, immunoprecipitation of HTRA1, a trypsin-like serine protease known to be secreted by cells differentiating into all three major mesenchymal lineages and by stressed cells in general and shown to degrade a number of matrix proteins including collagen, significantly diminished the homing effect. The data suggest that this protease is a major contributor to the observed chemotaxis of bone marrow stromal cells. The present study indicates that collagen fragments can mediate the migration of bone marrow stromal cells. The results also suggest that, at least in musculoskeletal and in adipose tissues, matrix remodeling occurrences, usually closely associated with tissue remodeling, should also be regarded as potential stem cells recruitment events.
Collapse
|
147
|
Huang HY, Hu LL, Song TJ, Li X, He Q, Sun X, Li YM, Lu HJ, Yang PY, Tang QQ. Involvement of cytoskeleton-associated proteins in the commitment of C3H10T1/2 pluripotent stem cells to adipocyte lineage induced by BMP2/4. Mol Cell Proteomics 2010; 10:M110.002691. [PMID: 20713452 DOI: 10.1074/mcp.m110.002691] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The developmental pathway that gives rise to mature adipocytes involves two distinct stages: commitment and terminal differentiation. Although the important proteins/factors contributing to terminal adipocyte differentiation have been well defined, the proteins/factors in the commitment of mesenchymal stem cells to the adipocyte lineage cells have not. In this study, we applied proteomics analysis profiling to characterize differences between uncommitted C3H10T1/2 pluripotent stem cells and those that have been committed to the adipocyte lineage by BMP4 or BMP2 with the goal to identify such proteins/factors and to understand the molecular mechanisms that govern the earliest stages of adipocyte lineage commitment. Eight proteins were found to be up-regulated by BMP2, and 27 proteins were up-regulated by BMP4, whereas five unique proteins were up-regulated at least 10-fold by both BMP2/4, including three cytoskeleton-associated proteins (i.e. lysyl oxidase (LOX), translationally controlled tumor protein 1 (TPT1), and αB-crystallin). Western blotting further confirmed the induction of the expression of these cytoskeleton-associated proteins in the committed C3H10T1/2 induced by BMP2/4. Importantly, knockdown of LOX expression totally prevented the commitment, whereas knockdown of TPT1 and αB-crystallin expression partially inhibited the commitment. Several published reports suggest that cell shape can influence the differentiation of partially committed precursors of adipocytes, osteoblasts, and chondrocytes. We observed a dramatic change of cell shape during the commitment process, and we showed that knockdown of these cytoskeleton-associated proteins prevented the cell shape change and restored F-actin organization into stress fibers and inhibited the commitment to the adipocyte lineage. Our studies indicate that these differentially expressed cytoskeleton-associate proteins might determine the fate of mesenchymal stem cells to commit to the adipocyte lineage through cell shape regulation.
Collapse
Affiliation(s)
- Hai-Yan Huang
- The Key Laboratory of Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai 200032, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
148
|
Cross talk between insulin and bone morphogenetic protein signaling systems in brown adipogenesis. Mol Cell Biol 2010; 30:4224-33. [PMID: 20584981 DOI: 10.1128/mcb.00363-10] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Both insulin and bone morphogenetic protein (BMP) signaling systems are important for adipocyte differentiation. Analysis of gene expression in BMP7-treated fibroblasts revealed a coordinated change in insulin signaling components by BMP7. To further investigate the cross talk between insulin and BMP signaling systems in brown adipogenesis, we examined the effect of BMP7 in insulin receptor substrate 1 (IRS-1)-deficient brown preadipocytes, which exhibit a severe defect in differentiation. Treatment of these cells with BMP7 for 3 days prior to adipogenic induction restored differentiation and expression of brown adipogenic markers. The high level of adipogenic inhibitor preadipocyte factor 1 (Pref-1) in IRS-1-null cells was markedly reduced by 3 days of BMP7 treatment, and analysis of the 1.3-kb pref-1 promoter revealed 9 putative Smad binding elements (SBEs), suggesting that BMP7 could directly suppress Pref-1 expression, thereby allowing the initiation of the adipogenic program. Using a series of sequential deletion mutants of the pref-1 promoter linked to the luciferase gene and chromatin immunoprecipitation, we demonstrate that the promoter-proximal SBE (-192/-184) was critical in mediating BMP7's suppressive effect on pref-1 transcription. Together, these data suggest cross talk between the insulin and BMP signaling systems by which BMP7 can rescue brown adipogenesis in cells with insulin resistance.
Collapse
|
149
|
Global comparison of gene expression profiles between intramuscular and subcutaneous adipocytes of neonatal landrace pig using microarray. Meat Sci 2010; 86:440-50. [PMID: 20573458 DOI: 10.1016/j.meatsci.2010.05.031] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2009] [Revised: 11/29/2009] [Accepted: 05/14/2010] [Indexed: 02/06/2023]
Abstract
The objective of this study was to compare the differences of gene expression profiles between intramuscular and subcutaneous adipocytes originated from the isolated preadipocytes in vitro. Cytosolic triglyceride determination indicated that subcutaneous adipocytes accumulated more lipid than intramuscular adipocytes did at the late stage of differentiation. Microarray assay revealed that 172 probes representing 133 genes were differentially expressed, among which 46 genes were highly expressed in intramuscular adipocytes and the other 87 genes were highly expressed in subcutaneous adipocytes. Real-time PCR confirmed that genes related to lipid metabolism, such as LPL, FABP4, FABP5 and OSBPL10, were predominantly expressed in subcutaneous adipocytes, whereas BMP4 and BMP7 were highly expressed in intramuscular adipocytes. The results indicated that the accumulation of lipid mass in subcutaneous adipocytes might be due to the highly expressed genes related to lipid metabolism, and the high levels of BMP4 and BMP7 in intramuscular adipocytes suggested that BMPs might be involved in the differentiation of intramuscular adipocytes.
Collapse
|
150
|
Ghosh S, Dean A, Walter M, Bao Y, Hu Y, Ruan J, Li R. Cell density-dependent transcriptional activation of endocrine-related genes in human adipose tissue-derived stem cells. Exp Cell Res 2010; 316:2087-98. [PMID: 20420826 DOI: 10.1016/j.yexcr.2010.04.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Revised: 03/23/2010] [Accepted: 04/19/2010] [Indexed: 10/19/2022]
Abstract
Adipose tissue is recognized as an endocrine organ that plays an important role in human diseases such as type II diabetes and cancer. Human adipose tissue-derived stem cells (ASCs), a distinct cell population in adipose tissue, are capable of differentiating into multiple lineages including adipogenesis. When cultured in vitro under a confluent condition, ASCs reach a commitment stage for adipogenesis, which can be further induced into terminally differentiated adipocytes by a cocktail of adipogenic factors. Here we report that the confluent state of ASCs triggers transcriptional activation cascades for genes that are responsible for the endocrine function of adipose tissue. These include insulin-like growth factor 1 (IGF-1) and aromatase (Cyp19), a key enzyme in estrogen biosynthesis. Despite similar adipogenic potentials, ASCs from different individuals display huge variations in activation of these endocrine-related genes. Bioinformatics and experimental data suggest that transcription factor Foxo1 controls a large number of "early" confluency-response genes, which subsequently induce "late" response genes. Furthermore, siRNA-mediated knockdown of Foxo1 substantially compromises the ability of committed ASCs to stimulate tumor cell migration in vitro. Thus, our work suggests that cell density is an important determinant of the endocrine potential of ASCs.
Collapse
Affiliation(s)
- Sagar Ghosh
- Department of Molecular Medicine, Institute of Biotechnology, 15355 Lambda Drive, University of Texas, Health Science Center at San Antonio, San Antonio, TX 78245, USA
| | | | | | | | | | | | | |
Collapse
|