1
|
Hashimoto T, Hirano K. Effects of mifepristone on adipocyte differentiation in mouse 3T3-L1 cells. Cell Mol Biol Lett 2024; 29:45. [PMID: 38553665 PMCID: PMC10981365 DOI: 10.1186/s11658-024-00559-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 02/29/2024] [Indexed: 04/01/2024] Open
Abstract
BACKGROUND Both glucocorticoid receptor and peroxisome proliferator-activated receptor-γ (PPARγ) play a critical role in adipocyte differentiation. Mifepristone is not only an antagonist of the glucocorticoid receptor but also an agonist of PPARγ. Therefore, the present study investigated the effect of mifepristone on adipocyte differentiation. METHODS Mouse 3T3-L1 cells were used as a model for adipocyte differentiation. The lipid droplet formation was evaluated with Bodipy493/503 staining and the expression of adipocyte markers [adiponectin and adipocyte fatty acid binding protein-4 (Fabp4)] was evaluated with quantitative PCR and immunoblot analyses for indication of adipocyte differentiation. siRNA and neutralizing antibodies were used to elucidate the molecular mechanism of mifepristone-induced adipocyte differentiation. Luciferase reporter assay was used to examine the effect of mifepristone on the promoter activity of PPAR-response element (PPRE). The DNA microarray analysis was used to characterize the transcriptome of the mifepristone-induced adipocytes. In vivo adipogenic effect of mifepristone was examined in mice. RESULTS Mifepristone not only enhanced adipocyte differentiation induced by the conventional protocol consisting of insulin, dexamethasone and 3-isobutyl-1-methylxanthine but also induced adipocyte differentiation alone, as evidenced by lipid droplets formation and induction of the expression of adiponectin and Fabp4. These effects were inhibited by an adiponectin-neutralizing antibody and a PPARγ antagonist. Mifepristone activated the promoter activity of PPRE in a manner sensitive to PPARγ antagonist. A principal component analysis (PCA) of DNA microarray data revealed that the mifepristone-induced adipocytes represent some characteristics of the in situ adipocytes in normal adipose tissues to a greater extent than those induced by the conventional protocol. Mifepristone administration induced an increase in the weight of epididymal, perirenal and gluteofemoral adipose tissues. CONCLUSIONS Mifepristone alone is capable of inducing adipocyte differentiation in 3T3-L1 cells and adipogenesis in vivo. PPARγ plays a critical role in the mifepristone-induced adipocyte differentiation. Mifepristone-induced adipocytes are closer to the in situ adipocytes than those induced by the conventional protocol. The present study proposes a single treatment with mifepristone as a novel protocol to induce more physiologically relevant adipocytes in 3T3-L1 cells than the conventional protocol.
Collapse
Affiliation(s)
- Takeshi Hashimoto
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University, 1750-1 Miki-Cho, Kita-Gun, Kagawa, 761-0793, Japan.
| | - Katsuya Hirano
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University, 1750-1 Miki-Cho, Kita-Gun, Kagawa, 761-0793, Japan
| |
Collapse
|
2
|
Zhao L, Jia D, Tan Z, Jiang H. Association of growth hormone deficiency with an increased number of preadipocytes in subcutaneous fat. Front Endocrinol (Lausanne) 2023; 14:1199589. [PMID: 37305046 PMCID: PMC10250704 DOI: 10.3389/fendo.2023.1199589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/10/2023] [Indexed: 06/13/2023] Open
Abstract
The inhibitory effect of growth hormone (GH) on adipose tissue growth is well known, but the underlying mechanism is not fully understood. In this study, we determined the possibility that GH inhibits adipose tissue growth by inhibiting adipogenesis, the process of formation of adipocytes from stem cells, in the lit/lit mice. The lit/lit mice are GH deficient because of a spontaneous mutation to the GH releasing hormone receptor (ghrhr) gene, and they have more subcutaneous fat despite being smaller than the lit/+ mice at the same age. We found that cells of the stromal vascular fraction (SVF) of subcutaneous fat from the lit/lit mice had greater adipogenic potential than those from the lit/+ mice, as evidenced by forming greater numbers of lipid droplets-containing adipocytes and having greater expression of adipocyte marker genes during induced adipocyte differentiation in culture. However, addition of GH to the culture did not reverse the superior adipogenic potential of subcutaneous SVF from the lit/lit mice. Through florescence-activated cell sorting and quantification of mRNAs of preadipocyte markers, including CD34, CD29, Sca-1, CD24, Pref-1, and PPARγ, we found that subcutaneous SVF from the lit/lit mice contained more preadipocytes than that from the lit/+ mice. These results support the notion that GH inhibits adipose tissue growth in mice at least in part by inhibiting adipogenesis. Furthermore, these results suggest that GH inhibits adipogenesis in mice not by inhibiting the terminal differentiation of preadipocytes into adipocytes, rather by inhibiting the formation of preadipocytes from stem cells or the recruitment of stem cells to the fat depot.
Collapse
|
3
|
Abstract
Worldwide obesity is increasing at an alarming rate in children and adolescents, with the consequent emergence of co-morbidities. Moreover, the maternal environment during pregnancy plays an important role in obesity, contributing to transgenerational transmission of the same and metabolic dysfunction. White adipose tissue represents a prime target of metabolic programming induced by maternal milieu. In this article, we review adipose tissue physiology and development, as well as maternal influences during the perinatal period that may lead to obesity in early postnatal life and adulthood. First, we describe the adipose tissue cell composition, distribution and hormonal action, together with the evidence of hormonal factors participating in fetal/postnatal programming. Subsequently, we describe the critical periods of adipose tissue development and the relationship of gestational and early postnatal life with healthy fetal adipose tissue expansion. Furthermore, we discuss the evidence showing that adipose tissue is an important target for nutritional, hormonal and epigenetic signals to modulate fetal growth. Finally, we describe nutritional, hormonal, epigenetic and microbiome changes observed in maternal obesity, and whether their disruption alters fetal growth and adiposity. The presented evidence supports the developmental origins of health and disease concept, which proposes that the homeostatic system is affected during gestational and postnatal development, impeding the ability to regulate body weight after birth, thereby resulting in adult obesity. Consequently, we anticipate that promoting a healthy early-life programming of adipose tissue and increasing the knowledge of the mechanisms by which maternal factors affect the health of future generations may offer novel strategies for explaining and addressing worldwide health problems such as obesity.
Collapse
|
4
|
Mohamed MYH, Christie G. A system for the expression and release of heterologous proteins from the core ofBacillus subtilisspores. FEMS Microbiol Lett 2018; 365:5195517. [DOI: 10.1093/femsle/fny270] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 11/20/2018] [Indexed: 12/15/2022] Open
Affiliation(s)
- Mohamed Y H Mohamed
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, UK
| | - Graham Christie
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, UK
| |
Collapse
|
5
|
Kuri-Harcuch W, Velez-delValle C, Vazquez-Sandoval A, Hernández-Mosqueira C, Fernandez-Sanchez V. A cellular perspective of adipogenesis transcriptional regulation. J Cell Physiol 2018; 234:1111-1129. [PMID: 30146705 DOI: 10.1002/jcp.27060] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 06/25/2018] [Indexed: 12/19/2022]
Abstract
Adipose cells store lipids in the cytoplasm and signal systemically through secretion of adipokines and other molecules that regulate body energy metabolism. Differentiation of fat cells and its regulation has been the focus of extensive research since the early 1970s. In this review, we had attempted to examine the research bearing on the control of adipose cell differentiation, some of it dating back to the early days when Howard Green and his group described the preadipocyte cell lines 3T3-L1 and 3T3-F442A during 1974-1975. We also concentrated our attention on research published during the last few years, emphasizing data described on transcription factors that regulate adipose differentiation, outside of those that were reported earlier as part of the canonical adipogenic transcriptional cascade, which has been the subject of ample reviews by several groups of researchers. We focused on the studies carried out with the two preadipocyte cell culture models, the 3T3-L1 and 3T3-F442A cells that have provided essential data on adipose biology.
Collapse
Affiliation(s)
- Walid Kuri-Harcuch
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Cristina Velez-delValle
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Alfredo Vazquez-Sandoval
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Claudia Hernández-Mosqueira
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Veronica Fernandez-Sanchez
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| |
Collapse
|
6
|
Olarescu NC, Berryman DE, Householder LA, Lubbers ER, List EO, Benencia F, Kopchick JJ, Bollerslev J. GH action influences adipogenesis of mouse adipose tissue-derived mesenchymal stem cells. J Endocrinol 2015; 226:13-23. [PMID: 25943560 PMCID: PMC4560118 DOI: 10.1530/joe-15-0012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/05/2015] [Indexed: 12/15/2022]
Abstract
GH influences adipocyte differentiation, but both stimulatory and inhibitory effects have been described. Adipose tissue-derived mesenchymal stem cells (AT-MSCs) are multipotent and are able to differentiate into adipocytes, among other cells. Canonical Wnt/β-catenin signaling activation impairs adipogenesis. The aim of the present study was to elucidate the role of GH on AT-MSC adipogenesis using cells isolated from male GH receptor knockout (GHRKO), bovine GH transgenic (bGH) mice, and wild-type littermate control (WT) mice. AT-MSCs from subcutaneous (sc), epididiymal (epi), and mesenteric (mes) AT depots were identified and isolated by flow cytometry (Pdgfrα+ Sca1+ Cd45- Ter119- cells). Their in vitro adipogenic differentiation capacity was determined by cell morphology and real-time RT-PCR. Using identical in vitro conditions, adipogenic differentiation of AT-MSCs was only achieved in the sc depot, and not in epi and mes depots. Notably, we observed an increased differentiation in cells isolated from sc-GHRKO and an impaired differentiation of sc-bGH cells as compared to sc-WT cells. Axin2, a marker of Wnt/β-catenin activation, was increased in mature sc-bGH adipocytes, which suggests that activation of this pathway may be responsible for the decreased adipogenesis. Thus, the present study demonstrates that (i) adipose tissue in mice has a well-defined population of Pdgfrα+ Sca1+ MSCs; (ii) the differentiation capacity of AT-MSCs varies from depot to depot regardless of GH genotype; (iii) the lack of GH action increases adipogenesis in the sc depot; and iv) activation of the Wnt/β-catenin pathway might mediate the GH effect on AT-MSCs. Taken together, the present results suggest that GH diminishes fat mass in part by altering adipogenesis of MSCs.
Collapse
Affiliation(s)
- Nicoleta C Olarescu
- Section of Specialized EndocrinologyDepartment of Endocrinology, Oslo University Hospital, Rikshospitalet, PO Box 4950, N-0424 Oslo, NorwayFaculty of MedicineUniversity of Oslo, Oslo, NorwayEdison Biotechnology InstituteOhio University, Athens, Ohio, USAHeritage College of Osteopathic MedicineOhio University, Athens, Ohio, USA Section of Specialized EndocrinologyDepartment of Endocrinology, Oslo University Hospital, Rikshospitalet, PO Box 4950, N-0424 Oslo, NorwayFaculty of MedicineUniversity of Oslo, Oslo, NorwayEdison Biotechnology InstituteOhio University, Athens, Ohio, USAHeritage College of Osteopathic MedicineOhio University, Athens, Ohio, USA
| | - Darlene E Berryman
- Section of Specialized EndocrinologyDepartment of Endocrinology, Oslo University Hospital, Rikshospitalet, PO Box 4950, N-0424 Oslo, NorwayFaculty of MedicineUniversity of Oslo, Oslo, NorwayEdison Biotechnology InstituteOhio University, Athens, Ohio, USAHeritage College of Osteopathic MedicineOhio University, Athens, Ohio, USA Section of Specialized EndocrinologyDepartment of Endocrinology, Oslo University Hospital, Rikshospitalet, PO Box 4950, N-0424 Oslo, NorwayFaculty of MedicineUniversity of Oslo, Oslo, NorwayEdison Biotechnology InstituteOhio University, Athens, Ohio, USAHeritage College of Osteopathic MedicineOhio University, Athens, Ohio, USA
| | - Lara A Householder
- Section of Specialized EndocrinologyDepartment of Endocrinology, Oslo University Hospital, Rikshospitalet, PO Box 4950, N-0424 Oslo, NorwayFaculty of MedicineUniversity of Oslo, Oslo, NorwayEdison Biotechnology InstituteOhio University, Athens, Ohio, USAHeritage College of Osteopathic MedicineOhio University, Athens, Ohio, USA Section of Specialized EndocrinologyDepartment of Endocrinology, Oslo University Hospital, Rikshospitalet, PO Box 4950, N-0424 Oslo, NorwayFaculty of MedicineUniversity of Oslo, Oslo, NorwayEdison Biotechnology InstituteOhio University, Athens, Ohio, USAHeritage College of Osteopathic MedicineOhio University, Athens, Ohio, USA
| | - Ellen R Lubbers
- Section of Specialized EndocrinologyDepartment of Endocrinology, Oslo University Hospital, Rikshospitalet, PO Box 4950, N-0424 Oslo, NorwayFaculty of MedicineUniversity of Oslo, Oslo, NorwayEdison Biotechnology InstituteOhio University, Athens, Ohio, USAHeritage College of Osteopathic MedicineOhio University, Athens, Ohio, USA
| | - Edward O List
- Section of Specialized EndocrinologyDepartment of Endocrinology, Oslo University Hospital, Rikshospitalet, PO Box 4950, N-0424 Oslo, NorwayFaculty of MedicineUniversity of Oslo, Oslo, NorwayEdison Biotechnology InstituteOhio University, Athens, Ohio, USAHeritage College of Osteopathic MedicineOhio University, Athens, Ohio, USA
| | - Fabian Benencia
- Section of Specialized EndocrinologyDepartment of Endocrinology, Oslo University Hospital, Rikshospitalet, PO Box 4950, N-0424 Oslo, NorwayFaculty of MedicineUniversity of Oslo, Oslo, NorwayEdison Biotechnology InstituteOhio University, Athens, Ohio, USAHeritage College of Osteopathic MedicineOhio University, Athens, Ohio, USA
| | - John J Kopchick
- Section of Specialized EndocrinologyDepartment of Endocrinology, Oslo University Hospital, Rikshospitalet, PO Box 4950, N-0424 Oslo, NorwayFaculty of MedicineUniversity of Oslo, Oslo, NorwayEdison Biotechnology InstituteOhio University, Athens, Ohio, USAHeritage College of Osteopathic MedicineOhio University, Athens, Ohio, USA Section of Specialized EndocrinologyDepartment of Endocrinology, Oslo University Hospital, Rikshospitalet, PO Box 4950, N-0424 Oslo, NorwayFaculty of MedicineUniversity of Oslo, Oslo, NorwayEdison Biotechnology InstituteOhio University, Athens, Ohio, USAHeritage College of Osteopathic MedicineOhio University, Athens, Ohio, USA
| | - Jens Bollerslev
- Section of Specialized EndocrinologyDepartment of Endocrinology, Oslo University Hospital, Rikshospitalet, PO Box 4950, N-0424 Oslo, NorwayFaculty of MedicineUniversity of Oslo, Oslo, NorwayEdison Biotechnology InstituteOhio University, Athens, Ohio, USAHeritage College of Osteopathic MedicineOhio University, Athens, Ohio, USA Section of Specialized EndocrinologyDepartment of Endocrinology, Oslo University Hospital, Rikshospitalet, PO Box 4950, N-0424 Oslo, NorwayFaculty of MedicineUniversity of Oslo, Oslo, NorwayEdison Biotechnology InstituteOhio University, Athens, Ohio, USAHeritage College of Osteopathic MedicineOhio University, Athens, Ohio, USA
| |
Collapse
|
7
|
Chia DJ. Minireview: mechanisms of growth hormone-mediated gene regulation. Mol Endocrinol 2014; 28:1012-25. [PMID: 24825400 DOI: 10.1210/me.2014-1099] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
GH exerts a diverse array of physiological actions that include prominent roles in growth and metabolism, with a major contribution via stimulating IGF-1 synthesis. GH achieves its effects by influencing gene expression profiles, and Igf1 is a key transcriptional target of GH signaling in liver and other tissues. This review examines the mechanisms of GH-mediated gene regulation that begin with signal transduction pathways activated downstream of the GH receptor and continue with chromatin events at target genes and additionally encompasses the topics of negative regulation and cross talk with other cellular inputs. The transcription factor, signal transducer and activator of transcription 5b, is regarded as the major signaling pathway by which GH achieves its physiological effects, including in stimulating Igf1 gene transcription in liver. Recent studies exploring the mechanisms of how activated signal transducer and activator of transcription 5b accomplishes this are highlighted, which begin to characterize epigenetic features at regulatory domains of the Igf1 locus. Further research in this field offers promise to better understand the GH-IGF-1 axis in normal physiology and disease and to identify strategies to manipulate the axis to improve human health.
Collapse
Affiliation(s)
- Dennis J Chia
- Department of Pediatrics, Icahn School of Medicine at Mt Sinai, New York, New York 10029
| |
Collapse
|
8
|
Kamenický P, Mazziotti G, Lombès M, Giustina A, Chanson P. Growth hormone, insulin-like growth factor-1, and the kidney: pathophysiological and clinical implications. Endocr Rev 2014; 35:234-81. [PMID: 24423979 DOI: 10.1210/er.2013-1071] [Citation(s) in RCA: 142] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Besides their growth-promoting properties, GH and IGF-1 regulate a broad spectrum of biological functions in several organs, including the kidney. This review focuses on the renal actions of GH and IGF-1, taking into account major advances in renal physiology and hormone biology made over the last 20 years, allowing us to move our understanding of GH/IGF-1 regulation of renal functions from a cellular to a molecular level. The main purpose of this review was to analyze how GH and IGF-1 regulate renal development, glomerular functions, and tubular handling of sodium, calcium, phosphate, and glucose. Whenever possible, the relative contributions, the nephronic topology, and the underlying molecular mechanisms of GH and IGF-1 actions were addressed. Beyond the physiological aspects of GH/IGF-1 action on the kidney, the review describes the impact of GH excess and deficiency on renal architecture and functions. It reports in particular new insights into the pathophysiological mechanism of body fluid retention and of changes in phospho-calcium metabolism in acromegaly as well as of the reciprocal changes in sodium, calcium, and phosphate homeostasis observed in GH deficiency. The second aim of this review was to analyze how the GH/IGF-1 axis contributes to major renal diseases such as diabetic nephropathy, renal failure, renal carcinoma, and polycystic renal disease. It summarizes the consequences of chronic renal failure and glucocorticoid therapy after renal transplantation on GH secretion and action and questions the interest of GH therapy in these conditions.
Collapse
Affiliation(s)
- Peter Kamenický
- Assistance Publique-Hôpitaux de Paris (P.K., M.L., P.C.), Hôpital de Bicêtre, Service d'Endocrinologie et des Maladies de la Reproduction, Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Le Kremlin Bicêtre F-94275, France; Univ Paris-Sud (P.K., M.L., P.C.), Faculté de Médecine Paris-Sud, Le Kremlin Bicêtre F-94276, France; Inserm Unité 693 (P.K., M.L., P.C.), Le Kremlin Bicêtre F-94276, France; and Department of Clinical and Experimental Sciences (A.G., G.M.), Chair of Endocrinology, University of Brescia, 25125 Brescia, Italy
| | | | | | | | | |
Collapse
|
9
|
Zhao L, Wang A, Corl BA, Jiang H. Effect of growth hormone on the differentiation of bovine preadipocytes into adipocytes and the role of the signal transducer and activator of transcription 5b. J Anim Sci 2014; 92:1958-67. [PMID: 24668956 DOI: 10.2527/jas.2013-7113] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We evaluated the effect of GH on the differentiation of primary bovine preadipocytes into adipocytes. Bovine preadipocytes, derived from adipose tissue explants, were induced to differentiate into adipocytes in the presence or absence of recombinant bovine GH. The differentiation status of adipocytes was assessed by Oil Red O staining and by measuring the activity of glycerol-3-phosphate dehydrogenase (G3PDH) and the rate of acetate incorporation. Fewer preadipocytes became adipocytes in the presence of GH than in the absence of GH; adipocytes formed in the presence of GH had lower G3PDH activity and lower rate of acetate incorporation than those formed without GH treatment (P < 0.05). These data suggest an inhibitory effect of GH on the differentiation of bovine preadipocytes into adipocytes. Growth hormone decreased the expression of C/EBPα and PPARγ mRNA in bovine adipocytes (P < 0.05). Because C/EBPα and PPARγ are the master regulators of adipocyte differentiation, this data suggests that GH might inhibit the differentiation of bovine preadipocytes into adipocytes by inhibiting the expression of C/EBPα and/or PPARγ. Because the signal transducer and activator of transcription 5 (STAT5) is a major component of signaling from the GH receptor, we next determined the potential role of STAT5 in GH inhibition of bovine adipocyte differentiation. Overexpression of a constitutively active form of STAT5b (STAT5bCA) in bovine preadipocytes through adenoviral transduction mimicked the effects of GH on the formation of lipid-containing adipocytes, G3PDH activity, and acetate incorporation rate. Overexpression of STAT5bCA was associated with decreased expression of C/EBPα mRNA (P < 0.05) but not that of PPARγ mRNA in bovine adipocytes. These results support a role of STAT5b in mediating GH inhibition of C/EBPα expression but not that of PPARγ expression in bovine preadipocytes. Overall, the present study suggests that GH may inhibit adipose growth in cattle in part by inhibiting adipogenesis and that GH inhibits the differentiation of bovine preadipocytes to adipocytes through STAT5b-dependent inhibition of C/EBPα expression and STAT5b-independent inhibition of PPARγ expression.
Collapse
Affiliation(s)
- L Zhao
- Department of Animal and Poultry Sciences
| | | | | | | |
Collapse
|
10
|
McMenamin SK, Minchin JEN, Gordon TN, Rawls JF, Parichy DM. Dwarfism and increased adiposity in the gh1 mutant zebrafish vizzini. Endocrinology 2013; 154:1476-87. [PMID: 23456361 PMCID: PMC3602633 DOI: 10.1210/en.2012-1734] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Somatic growth and adipogenesis are closely associated with the development of obesity in humans. In this study, we identify a zebrafish mutant, vizzini, that exhibits both a severe defect in somatic growth and increased accumulation of adipose tissue. Positional cloning of vizzini revealed a premature stop codon in gh1. Although the effects of GH are largely through igfs in mammals, we found no decrease in the expression of igf transcripts in gh1 mutants during larval development. As development progressed, however, we found overall growth to be progressively retarded and the attainment of specific developmental stages to occur at abnormally small body sizes relative to wild type. Moreover, both subcutaneous (sc) and visceral adipose tissues underwent precocious development in vizzini mutants, and at maturity, the sizes of different fat deposits were greatly expanded relative to wild type. In vivo confocal imaging of sc adipose tissue (SAT) expansion revealed that vizzini mutants exhibit extreme enlargement of adipocyte lipid droplets without a corresponding increase in lipid droplet number. These findings suggest that GH1 signaling restricts SAT hypertrophy in zebrafish. Finally, nutrient deprivation of vizzini mutants revealed that SAT mobilization was greatly diminished during caloric restriction, further implicating GH1 signaling in adipose tissue homeostasis. Overall, the zebrafish gh1 mutant, vizzini, exhibits decreased somatic growth, increased adipose tissue accumulation, and disrupted adipose plasticity after nutrient deprivation and represents a novel model to investigate the in vivo dynamics of vertebrate obesity.
Collapse
Affiliation(s)
- Sarah K McMenamin
- Department of Biology, University of Washington, Box 351800, Seattle, Washington 98195-1800, USA
| | | | | | | | | |
Collapse
|
11
|
Takenouchi T, Takayama Y, Takezawa T. Co-treatment with dexamethasone and octanoate induces adipogenesis in 3T3-L1 cells. Cell Biol Int 2013; 28:209-16. [PMID: 14984747 DOI: 10.1016/j.cellbi.2003.11.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2003] [Revised: 10/10/2003] [Accepted: 11/27/2003] [Indexed: 11/24/2022]
Abstract
We report here that octanoate, a medium chain fatty acid, induces adipocyte differentiation in 3T3-L1 cells by co-treatment with dexamethasone, although octanoate has been known not to stimulate 3T3-L1 adipogenesis. A low concentration of exogenous glucose prevented 3T3-L1 adipogenesis induced by 1-methyl 3-isobutylxanthine, dexamethasone, and insulin (MDI) treatment (a common protocol for adipocyte differentiation). In contrast, co-treatment with dexamethasone and octanoate (D-OCT) induced adipogenesis under the same conditions. These findings imply that octanoate, rather than glucose, is the source of accumulated lipids in D-OCT-induced adipogenesis. D-OCT increased expression of the differentiation markers peroxisome proliferator-activated receptor (PPAR)gamma2 and caveolin-1. A specific inhibitor of p38 mitogen-activated protein (MAP) kinase inhibited D-OCT-induced adipogenesis. These results suggest that the p38 MAP kinase pathway followed by up-regulation of PPARgamma2 may be involved in 3T3-L1 adipocyte differentiation induced by D-OCT, as well as by MDI.
Collapse
Affiliation(s)
- Takato Takenouchi
- Functional Biomolecule Laboratory, National Institute of Livestock and Grassland Science, Ikenodai 2, Tsukuba, Ibaraki 305-0901, Japan.
| | | | | |
Collapse
|
12
|
Affiliation(s)
- Jeff Holly
- Academic Units of Surgery and Child Health, University of Bristol, Bristol, United Kingdom
| | | | | | | |
Collapse
|
13
|
Flynn L, Woodhouse KA. Adipose tissue engineering with cells in engineered matrices. Organogenesis 2012; 4:228-35. [PMID: 19337402 DOI: 10.4161/org.4.4.7082] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2008] [Accepted: 09/29/2008] [Indexed: 12/20/2022] Open
Abstract
Tissue engineering has shown promise for the development of constructs to facilitate large volume soft tissue augmentation in reconstructive and cosmetic plastic surgery. This article reviews the key progress to date in the field of adipose tissue engineering. In order to effectively design a soft tissue substitute, it is critical to understand the native tissue environment and function. As such, the basic physiology of adipose tissue is described and the process of adipogenesis is discussed. In this article, we have focused on tissue engineering using a cell-seeded scaffold approach, where engineered extracellular matrix substitutes are seeded with exogenous cells that may contribute to the regenerative response. The strengths and limitations of each of the possible cell sources for adipose tissue engineering, including adipose-derived stem cells, are detailed. We briefly highlight some of the results from the major studies to date, involving a range of synthetic and naturally derived scaffolds. While these studies have shown that adipose tissue regeneration is possible, more research is required to develop optimized constructs that will facilitate safe, predictable and long-term augmentation in clinical applications.
Collapse
Affiliation(s)
- Lauren Flynn
- Department of Chemical Engineering; Queen's University; Ontario Canada
| | | |
Collapse
|
14
|
Aoki N, Kawada T, Sugimoto E. Level of Preadipocyte Growth Factor in Rat Adipose Tissue which Specifically Permits the Proliferation of Preadipocytes Is Affected by Restricted Energy Intake. ACTA ACUST UNITED AC 2012; 1:126-31. [PMID: 16350568 DOI: 10.1002/j.1550-8528.1993.tb00601.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We previously reported the presence of a protein growth factor in rat adipose tissue which specifically permits the proliferation of 3T3-L1 and Ob1771 preadipocytes [Biochem. Biophys. Res. Commun. 1990;171:905-912, ref. 1] and which is hereinafter referred to as PAGF (preadipocyte growth factor). In this study, the effects of long-term restricted energy intake on the PAGF activity in rat epididymal and perirenal adipose tissue toward 3T3-L1 preadipocytes were investigated. When rats were subjected to restricted energy intake for three weeks, PAGF activity increased with energy intake. The body weight, epididymal and perirenal fat depot weights and glycerol 3-phosphate dehydrogenase activity also increased with the energy intake, whereas the lactate dehydrogenase activity remained almost constant in all energy intake groups. These results suggest that the PAGF in fat depots functions in response to energy intake and contributes to the de novo formation of adipocytes and the growth of adipose tissue. This factor may provide a useful tool for further elucidation of the relationship between energy storage in adipose tissue and adipose tissue development.
Collapse
Affiliation(s)
- N Aoki
- Laboratory of Nutritional Chemistry, Dept. of Food Science and Tech., School of Agriculture, Nagoya University, Nagoya, Aichi 464-01, Japan
| | | | | |
Collapse
|
15
|
Garten A, Schuster S, Kiess W. The insulin-like growth factors in adipogenesis and obesity. Endocrinol Metab Clin North Am 2012; 41:283-95, v-vi. [PMID: 22682631 DOI: 10.1016/j.ecl.2012.04.011] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Adipose tissue has been recognized as a major target of growth hormone (GH) action. GH was shown to inhibit adipocyte differentiation but stimulated preadipocyte proliferation in vitro. GH acts directly via its receptor or via upregulating insulin-like growth factor (IGF)-I, which is a critical mediator of preadipocyte proliferation, differentiation, and survival. Results from clinical studies on GH treatment in patients with GH deficiency or GH insensitivity syndrome can be used to dissect GH and IGF as well as IGF-binding protein (IGFBP) actions in vivo. In this article, changes of the GH/IGF system during adipocyte differentiation in vitro as well as related signaling pathways and their impact on adipose tissue growth and function are discussed. Clinical considerations include the effects of GH and IGF-I on adipose tissue during treatment of GH deficiency, differences in the IGF system between visceral and subcutaneous adipose tissue depots as well as the recently emerging role for adipose tissue in the regulation of glucose homeostasis.
Collapse
Affiliation(s)
- A Garten
- Department of Women and Child Health, Hospital for Children and Adolescents, Center for Pediatric Research Leipzig, University Hospitals, Liebigstraße 20a, 04103 Leipzig, Germany
| | | | | |
Collapse
|
16
|
|
17
|
KYOYA T, ISHIDA A, NAKASHIMA K, NAKAJIMA I, TOYODA A, NAKAMURA Y, KATSUMATA M. The effects of concentrations of lysine in media on differentiation of 3T3-L1 preadipocytes. Anim Sci J 2011; 82:565-70. [DOI: 10.1111/j.1740-0929.2011.00882.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
18
|
Smit LS, Meyer DJ, Argetsinger LS, Schwartz J, Carter‐Su C. Molecular Events in Growth Hormone–Receptor Interaction and Signaling. Compr Physiol 2011. [DOI: 10.1002/cphy.cp070514] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
19
|
Lu C, Kumar PA, Fan Y, Sperling MA, Menon RK. A novel effect of growth hormone on macrophage modulates macrophage-dependent adipocyte differentiation. Endocrinology 2010; 151:2189-99. [PMID: 20185763 PMCID: PMC2869256 DOI: 10.1210/en.2009-1194] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The GH receptor (GHR) is expressed on macrophages. However, the precise role of GH in regulation of macrophage function is unclear. We hypothesized that soluble factors including cytokines produced by macrophages in a GH-dependent manner regulate adipogenesis. We confirmed expression and functional integrity of the GHR in the J774A.1 macrophage cells. Conditioned medium (CM) from macrophages inhibited adipogenesis in a 3T3-L1 adipogenesis assay. CM from GH-treated macrophages decreased the inhibitory effect of CM from macrophages on adipogenesis. This effect on preadipocyte differentiation was active only during the first (early) phase of adipocyte differentiation. CM from stromal vascular compartment macrophages of mice with macrophage-specific deletion of the GHR exhibited more inhibitory effect on 3T3-L1 preadipocyte differentiation compared with CM from stromal vascular compartment macrophages of control mice, indicating that intact GH action in primary macrophages also increases preadipocyte differentiation. GH did not increase IGF-1 expression in macrophages. PCR array analysis identified IL-1beta as a candidate cytokine whose expression was altered by GH in macrophages. Levels of IL-1beta mRNA and protein were significantly decreased in GH-treated J774A.1 macrophages. Nuclear factor-kappaB stimulates IL-1beta gene expression, and GH induced a significant decrease in the levels of phosphorylated nuclear factor-kappaB in macrophages. IL-1beta is a known inhibitor of adipogenesis, and these results support GH-dependent down-regulation of macrophage IL-1beta expression as one mechanism for the observed increase in adipogenesis with CM from GH-treated macrophages. We conclude that GH decreases secretion of IL-1beta by the macrophage and thus in a paracrine manner increases adipocyte differentiation. These results provide a novel mechanism for GH's actions in the control of adipogenesis.
Collapse
Affiliation(s)
- Chunxia Lu
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan 48109-0718, USA
| | | | | | | | | |
Collapse
|
20
|
Frare EO, Santello FH, Caetano LC, Caldeira JC, Toldo MPA, Prado JCD. Growth hormones therapy in immune response against Trypanosoma cruzi. Res Vet Sci 2010; 88:273-8. [DOI: 10.1016/j.rvsc.2009.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2009] [Revised: 08/10/2009] [Accepted: 10/03/2009] [Indexed: 10/20/2022]
|
21
|
Barrera-Saldaña H, Ascacio-Martínez J, Sifuentes-Rincón A, Arellano-Vera W, Arbiza S. Applications of biotechnology and genomics in goats. Small Rumin Res 2010. [DOI: 10.1016/j.smallrumres.2009.12.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
22
|
Chin HS, Pack KJ, Pack SH, Kim JK. The Effects of Herbal Extract Mixture on Anti-Obesity. ACTA ACUST UNITED AC 2009. [DOI: 10.3746/jkfn.2009.38.1.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
23
|
Diaz-Velasquez CE, Castro-Muñozledo F, Kuri-Harcuch W. Staurosporine rapidly commits 3T3-F442A cells to the formation of adipocytes by activation of GSK-3β and mobilization of calcium. J Cell Biochem 2008; 105:147-57. [DOI: 10.1002/jcb.21810] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
24
|
Jin H, Lanning NJ, Carter-Su C. JAK2, but not Src family kinases, is required for STAT, ERK, and Akt signaling in response to growth hormone in preadipocytes and hepatoma cells. Mol Endocrinol 2008; 22:1825-41. [PMID: 18499741 DOI: 10.1210/me.2008-0015] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Janus kinase 2 (JAK2), a tyrosine kinase that associates with the GH receptor and is activated by GH, has been implicated as a key mediator of GH signaling. Several published reports suggest that members of the Src family of tyrosine kinases may also participate in GH signaling. We therefore investigated the extent to which JAK2 and Src family kinases mediate GH activation of signal transducers and activators of transcription (STATs) 1, 3, and 5a/b, ERKs 1 and 2, and Akt, in the highly GH-responsive cell lines 3T3-F442A preadipocytes and H4IIE hepatoma cells. GH activation of Src family kinases was not detected in either cell line. Further, blocking basal activity of Src kinases with the Src inhibitors PP1 and PP2 did not inhibit GH activation of STATs 1, 3, or 5a/b, or ERKs 1 and 2. When levels of JAK2 were depressed by short hairpin RNA in 3T3-F442A and H4IIE cells, GH-stimulated activation of STATs 1, 3, and 5a/b, ERKs 1 and 2, and Akt were significantly reduced; however, basal activity of Src family kinases was unaffected. These results were supported genetically by experiments showing that GH robustly activates JAK2, STATs 3 and 5a/b, ERKs 1 and 2, and Akt in murine embryonic fibroblasts derived from Src/Yes/ Fyn triple-knockout embryos that lack known Src kinases. These results strongly suggest that JAK2, but not Src family kinases, is critical for transducing these GH signals in 3T3-F442A and H4IIE cells.
Collapse
Affiliation(s)
- Hui Jin
- Department of Molecular and Integrative Physiology, The University of Michigan Medical School, Ann Arbor, Michigan 48109-5622, USA
| | | | | |
Collapse
|
25
|
McElvaine AT, Korytko AI, Kilen SM, Cuttler L, Mayo KE. Pituitary-Specific Expression and Pit-1 Regulation of the Rat Growth Hormone-Releasing Hormone Receptor Gene. Mol Endocrinol 2007; 21:1969-83. [PMID: 17536003 DOI: 10.1210/me.2007-0116] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The GHRH receptor is expressed in the somatotroph cell of the anterior pituitary, where it functions to mediate GHRH-stimulated GH release. To study pituitary and somatotroph cell-specific expression of this gene, a transgenic mouse model and complementary cell culture experiments were developed. The activity of the 1.6-kb proximal rat GHRH receptor promoter was examined in vivo by generating transgenic mice with the promoter directing expression of a luciferase reporter. The promoter directs tissue-specific expression; luciferase is highly expressed in the pituitary but absent from 14 other tissues. Immunocytochemistry experiments show that transgene expression is targeted to GH-expressing somatotroph cells. The transgene is 5-fold more highly expressed in males than females, and there is an increase in transgene expression leading up to the onset of puberty. The 1.6-kb promoter was further examined in cell culture experiments, which revealed that the promoter is selectively activated in pituitary cells and that promoter-reporter expression in nonpituitary cells can be enhanced by the pituitary-specific transcription factor Pit-1. EMSAs identified 10 short regions that specifically bind Pit-1 with highly variable relative affinities. The highest affinity site was previously identified and is required for Pit-1 activation of the promoter. Four additional sites contribute to Pit-1 regulation of the promoter and are important to achieving full activation of the gene. The results show that the 1.6-kb promoter is sufficient to direct tissue- and cell-specific expression in vivo and is regulated by Pit-1.
Collapse
Affiliation(s)
- Allison T McElvaine
- Department of Biochemistry, Molecular Biology, and Cell Biology, Northwestern University, Hogan 4-112, Evanston, Illinois 60208, USA
| | | | | | | | | |
Collapse
|
26
|
Ishizeki K, Kagiya T, Fujiwara N, Harada H. In vitro adipocytic conversion in Meckel's chondrocytes in response to a fatty acid-containing medium. ARCHIVES OF HISTOLOGY AND CYTOLOGY 2006; 69:163-71. [PMID: 17031022 DOI: 10.1679/aohc.69.163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Chick serum (CKS) contains factors that stimulate adipocytes in Meckel's chondrocytes in vitro. In the present study, we analyzed levels of fatty acids in CKS, and further examined whether these had the potential to convert chondrocytes to adipocytes. Phenotypic changes were evaluated by light and electron microscopies, bromodeoxyuridine (BrdU) incorporation, triglyceride assays, and immunocytochemistry. We showed that CKS contained high levels of fatty acids, and a mixed medium containing 5 particular fatty acids inhibited DNA synthesis and the proliferation of chondrocytes as it facilitated their differentiation into adipocytes. The adipocytes produced were sudan-positive multilocular cells that morphologically and histochemically resembled adipocytes induced by the CKS-containing medium. Almost all lipid droplet-containing cells were positive for leptin and alpha-glycerophosphate dehydrogenase (GPDH), as evaluated by immunoperoxidase staining, and their triglyceride concentrations markedly increased during 4 to 6 days of culture. These results suggested that specific fatty acids in CKS are involved in the adipocytic conversion of Meckel's chondrocytes.
Collapse
Affiliation(s)
- Kiyoto Ishizeki
- Department of Oral Anatomy, School of Dentistry, Iwate Medical University, Morioka, Japan.
| | | | | | | |
Collapse
|
27
|
Blüher S, Kratzsch J, Kiess W. Insulin-like growth factor I, growth hormone and insulin in white adipose tissue. Best Pract Res Clin Endocrinol Metab 2005; 19:577-87. [PMID: 16311218 DOI: 10.1016/j.beem.2005.07.011] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Maturation of adipose tissue results from both the expansion of mature adipocytes and the formation of new adipocytes from adipocyte precursor cells. A variety of hormones related to adipogenesis have been identified recently. Both growth hormone (GH) and insulin-like growth factor I (IGF-I) are of major significance in adipocyte differentiation. IGF-I has been suggested to be a major regulator of cell proliferation, differentiation and metabolism, thus regulating, among other biological processes, adipose tissue growth and differentiation of pre-adipocytes into adipocytes. GH exerts its effects by increasing the pool of adipocyte precursor cells capable of differentiating into mature adipocytes. In addition, GH seems to have the potential to reduce the volume of mature adipocytes, thus inhibiting the expansion of adipose tissue and reducing body fat. This chapter gives an overview of studies that have investigated the roles of insulin, GH and IGF-I in adipogenesis.
Collapse
Affiliation(s)
- Susann Blüher
- Hospital for Children and Adolescents, University of Leipzig, Oststrasse 21-25, Germany
| | | | | |
Collapse
|
28
|
Louveau I, Gondret F. Regulation of development and metabolism of adipose tissue by growth hormone and the insulin-like growth factor system. Domest Anim Endocrinol 2004; 27:241-55. [PMID: 15451072 DOI: 10.1016/j.domaniend.2004.06.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2004] [Accepted: 06/21/2004] [Indexed: 01/19/2023]
Abstract
White adipose tissue plays a key role in the regulation of the energy balance of vertebrates. This tissue is also now recognized to secrete a variety of factors such as leptin, which is thought to be involved in the modulation of adipose mass. Unlike other tissues, adipose tissue mass has considerable capacity to expand. The review deals primarily on the regulation of development and metabolism of adipose tissue by growth hormone (GH) and the insulin-like growth factor (IGF) system, with a special focus on the pig. The anti-insulin effects of GH are well-documented in pigs as in other species. In vitro exposure of adipose precursor cells to GH leads to a decrease in differentiation of those cells in pigs, in contrast to data obtained in murine cell lines. In vivo treatment and prolonged in vitro incubation of adipose tissue or isolated adipocytes with GH result in a decrease in glucose transport and lipogenesis, especially at the level of the fatty acid synthase gene, resulting in a reduction of the lipid content and adipose tissue mass. The mechanism by which GH antagonizes insulin stimulation of lipogenesis is still unresolved, as it is not mediated by protein kinase A, protein kinase C and Janus kinase-2 at the signaling level, or upstream stimulatory factor 1 or sterol regulatory element binding protein-1 at the transcriptional level. GH is apparently the main regulator of IGF-I mRNA expression in adipose tissue, however, the effects of IGF-I on this tissue are rather unclear.
Collapse
Affiliation(s)
- I Louveau
- Institut National de la Recherche Agronomique, Unité Mixte de Recherches sur le Veau et le Porc, 35590 Saint Gilles, France.
| | | |
Collapse
|
29
|
Abstract
The use of experimental models is the foundation of experimental biology, so it is important to know how much the models can tell us about actual animals. Inconsistent or contradictory results from in vitro models are often associated with the perception that a particular model or results are somehow wrong and therefore cannot tell us anything important about how an animal works. In fact, in vitro conditions do not create new biology. Differences between in vitro and in vivo behavior can only result from the actual cellular repertoire, which provides a powerful tool to uncover new information. Adipose tissue research provides a useful context for examining this issue because the regulation of adipose growth and metabolism has important economic implications for livestock production. Examples are discussed in which either excess skepticism or narrow interpretation of results slowed progress toward our current understanding of adipose biology. Similarly, contemporary examples using genomics are used to suggest that large inconsistencies are still apparent with in vitro methods. Careful consideration of these inconsistencies may provide new insights.
Collapse
Affiliation(s)
- J Novakofski
- Department of Animal Sciences, University of Illinois, Urbana 61801, USA.
| |
Collapse
|
30
|
Ishizeki K, Takahashi N, Nawa T. Induction of adipogenesis by the intrasplenic transplantation of chick serum clots. ACTA ACUST UNITED AC 2004; 67:21-30. [PMID: 15125020 DOI: 10.1679/aohc.67.21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Chick serum contains a factor that stimulates adipogenesis in Meckel's chondrocytes in vitro. The present study examined whether chick serum has a capacity for adipogenic induction in vivo, by transplanting serum clots (created by drying chick serum for up to 4 weeks) into mouse spleens. Specimens were harvested for histological analyses, which included light and electron microscopy and immunohistochemistry. The transplanted serum clots induced the appearance of lipid droplet-containing cells in splenic cords and sinus. Almost all the lipid droplet-containing cells were positive for sudan staining and consisted of multilocular lipid vacuoles. Immunostaining showed that the adipocytes induced by transplantation of the serum clots initially appeared as peroxisome proliferator-activated receptor-gamma (PPARgamma)-positive cells and developed into leptin and alpha-glycerophosphate dehydrogenase (GPDH)-producing cells, in addition to type III collagen synthesis. Furthermore, double immunofluorescence staining revealed that the immunoreactivity for GPDH was detected not only in stromal cells but also in macrophages. It was thus confirmed that stromal cells and macrophages in the spleen contain lipid droplets as seen in intact white adipose cells. The present results suggest that chick serum contains factors for adipocyte induction not only in vitro but also in vivo, and that the adipogenic potential does not depend on the supplements used during the cell culture.
Collapse
Affiliation(s)
- Kiyoto Ishizeki
- Department of Oral Anatomy, School of Dentistry, Iwate Medical University, Morioka, Japan.
| | | | | |
Collapse
|
31
|
Iida K, Takahashi Y, Kaji H, Yoshioka S, Murata M, Iguchi G, Okimura Y, Chihara K. Diverse regulation of full-length and truncated growth hormone receptor expression in 3T3-L1 adipocytes. Mol Cell Endocrinol 2003; 210:21-9. [PMID: 14615057 DOI: 10.1016/j.mce.2003.08.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Two truncated forms of growth hormone (GH) receptor (GHR), 1-277 and 1-279, were reported to be normally produced in human tissues by alternative splicing in exon 9 and its boundary. We found previously that GHR-277 exerts a dominant-negative effect on full-length GHR (GHR-fl)-mediated GH signaling causing short stature. The existence of truncated GHRs (hGHR-tr) in normal tissues suggests that hGHR-tr may play a physiological role in regulation of GH action at the cellular level. To clarify the physiological significance of GHR-tr and the regulation mechanism of GHR-tr expression, we examined the expression of mouse GHR-tr (mGHR-tr) mRNA in mouse adipocyte 3T3-L1 cells, comparing with that of mouse GHR-fl (mGHR-fl). The mRNAs of two mGHR-tr, mGHR-282 and mGHR-280, were detected by RT-PCR methods using specific primers. Although the mGHR-282 and mGHR-280 mRNA levels were approximately 100 times lower than that of mGHR-fl in mature 3T3-L1 cells, quantitative analysis by competitive RT-PCR methods revealed that the mRNA levels of mGHR-280 in 3T3-L1 cells were transiently reduced and thereafter increased during differentiation from preadipocyte to adipocyte. In contrast, the mRNA levels of mGHR-fl were increased in parallel with the progress of differentiation. Stimulation by GH of differentiated 3T3-L1 mature adipocytes resulted in dose-dependent increases of the mRNA of both mGHR-fl and mGHR-282, whereas it caused a paradoxical decrease of the mRNA of mGHR-280 stimulated by high concentration of GH. These findings suggest that the expressions of truncated mGHRs were regulated in a different manner from that of mGHR-fl, thereby modulating GH action in murine adipocytes.
Collapse
Affiliation(s)
- Keiji Iida
- Division of Endocrinology/Metabolism, Neurology and Hematology/Oncology, Department of Clinical Molecular Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Juárez-Aguilar E, Castro-Muñozledo F, Kuri-Harcuch W. A simple and sensitive assay for GH activity based on 3T3-F442A cell differentiation. Biochem Biophys Res Commun 2003; 311:935-41. [PMID: 14623271 DOI: 10.1016/j.bbrc.2003.10.090] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We describe a fast, sensitive, specific, and simple in vitro assay for GH biological activity, based on the differentiation of 3T3-F442A cells into adipocytes. The 3T3-F442A cells were directly plated at 1.5 x 10(4)cells/cm(2) in medium with or without various concentrations of human growth hormone (hGH). After 7 days, cells were lysed with buffer containing 0.5 % (v/v) Triton X-100, and adipose conversion was quantitated by the activity of the adipogenic enzyme glycerophosphate dehydrogenase. The assay is highly sensitive and specific for GH from different species. These culture conditions have shortened the time for the cells to undergo adipose differentiation, and they might also be useful to design and test drugs or agents that modify adipocyte differentiation or lipid metabolism, or for evaluation of cytotoxic and pharmacologic effects of drugs and other compounds.
Collapse
Affiliation(s)
- Enrique Juárez-Aguilar
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del I.P.N., Apdo. Postal 14-740, 07000, México, D.F., Mexico
| | | | | |
Collapse
|
33
|
Piwien Pilipuk G, Galigniana MD, Schwartz J. Subnuclear localization of C/EBP beta is regulated by growth hormone and dependent on MAPK. J Biol Chem 2003; 278:35668-77. [PMID: 12821655 DOI: 10.1074/jbc.m305182200] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Localization of transcription regulatory proteins in the nucleus is dynamically regulated, and may alter nucleoplasmic concentrations and/or assembly of multimolecular transcription regulatory complexes, which ultimately regulate gene expression. Since growth hormone (GH) regulates multiple transcription factors including C/EBP beta, the effect of GH on the subcellular localization of C/EBP beta was examined in 3T3-F442A preadipocytes. Indirect immunofluorescence shows that C/EBP beta is diffusely distributed in nuclei of quiescent cells. Within 5 min of GH treatment, the diffuse pattern dramatically becomes punctate. The relocalization of C/EBP beta coincides with DAPI staining of heterochromatin. Further, C/EBP beta and heterochromatin protein (HP)-1 alpha colocalize in the nucleus, consistent with localization of C/EBP beta to pericentromeric heterochromatin. In contrast, C/EBP delta exhibits a diffuse distribution in the nucleus that is not modified by GH treatment. C/EBP beta is rapidly and transiently phosphorylated on a conserved MAPK consensus site in response to GH (Piwien-Pilipuk, G., MacDougald, O. A., and Schwartz, J. (2002) J. Biol. Chem. 277, 44557-44565). Indirect immunofluorescence using antibodies specific for C/EBP beta phosphorylated on the conserved MAPK site shows that GH also rapidly induces a punctate pattern of staining for the phosphorylated C/EBP beta. In addition, phosphorylated C/EBP beta colocalizes to pericentromeric heterochromatin. The satellite DNA present in heterochromatin contains multiple C/EBP binding sites. DNA binding analysis shows that C/EBP beta, C/EBP delta, and C/EBP alpha (p42 and p30 forms) can bind to satellite DNA as homo- or heterocomplexes in vitro. Importantly, GH rapidly and transiently increases binding of endogenous C/EBP beta from 3T3-F442A cells to satellite DNA. Further, the GH-promoted nuclear relocalization of C/EBP beta to pericentromeric heterochromatin was prevented by the MEK inhibitor U0126. This observation suggests that GH-dependent MAPK activation plays a role in the regulation of nuclear relocalization of C/EBP beta. Nuclear redistribution introduces a new level of transcriptional regulation in GH action, since GH-mediated phosphorylation and nuclear redistribution of C/EBP beta may be coordinated to achieve spatial-temporal control of gene expression.
Collapse
Affiliation(s)
- Graciela Piwien Pilipuk
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | |
Collapse
|
34
|
Scacchi M, Pincelli AI, Cavagnini F. Nutritional status in the neuroendocrine control of growth hormone secretion: the model of anorexia nervosa. Front Neuroendocrinol 2003; 24:200-24. [PMID: 14596812 DOI: 10.1016/s0091-3022(03)00014-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Growth hormone (GH) plays a key role not only in the promotion of linear growth but also in the regulation of intermediary metabolism, body composition, and energy expenditure. On the whole, the hormone appears to direct fuel metabolism towards the preferential oxidation of lipids instead of glucose and proteins, and to convey the energy derived from metabolic processes towards the synthesis of proteins. On the other hand, body energy stores and circulating energetic substrates take an important part in the regulation of somatotropin release. Finally, central and peripheral peptides participating in the control of food intake and energy expenditure (neuropeptide Y, leptin, and ghrelin) are also involved in the regulation of GH secretion. Altogether, nutritional status has to be regarded as a major determinant in the regulation of the somatotropin-somatomedin axis in animals and humans. In these latter, overweight is associated with marked impairment of spontaneous and stimulated GH release, while acute dietary restriction and chronic undernutrition induce an amplification of spontaneous secretion together with a clear-cut decrease in insulin-like growth factor I (IGF-I) plasma levels. Thus, over- and undernutrition represent two conditions connoted by GH hypersensitivity and GH resistance, respectively. Anorexia nervosa (AN) is a psychiatric disorder characterized by peculiar changes of the GH-IGF-I axis. In these patients, low circulating IGF-I levels are associated with enhanced GH production rate, highly disordered mode of somatotropin release, and variability of GH responsiveness to different pharmacological challenges. These abnormalities are likely due not only to the lack of negative IGF-I feedback, but also to a primary hypothalamic alteration with increased frequency of growth hormone releasing hormone discharges and decreased somatostatinergic tone. Given the reversal of the above alterations following weight recovery, these abnormalities can be seen as secondary, and possibly adaptive, to nutritional deprivation. The model of AN may provide important insights into the pathophysiology of GH secretion, in particular as regards the mechanisms whereby nutritional status effects its regulation.
Collapse
Affiliation(s)
- Massimo Scacchi
- Chair of Endocrinology, University of Milan, Ospedale San Luca IRCCS, Istituto Auxologico Italiano, Milan, Italy
| | | | | |
Collapse
|
35
|
Zhang H, Nøohr J, Jensen CH, Petersen RK, Bachmann E, Teisner B, Larsen LK, Mandrup S, Kristiansen K. Insulin-like growth factor-1/insulin bypasses Pref-1/FA1-mediated inhibition of adipocyte differentiation. J Biol Chem 2003; 278:20906-14. [PMID: 12651852 DOI: 10.1074/jbc.m300022200] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Pref-1 is a highly glycosylated Delta-like transmembrane protein containing six epidermal growth factor-like repeats in the extracellular domain. Pref-1 is abundantly expressed in preadipocytes, but expression is down-regulated during adipocyte differentiation. Forced expression of Pref-1 in 3T3-L1 cells was reported to inhibit adipocyte differentiation. Here we show that efficient and regulated processing of Pref-1 occurs in 3T3-L1 preadipocytes releasing most of the extracellular domain as a 50-kDa heterogeneous protein, previously isolated and characterized as FA1. Unexpectedly, we found that forced expression of the soluble form, FA1, or full-length Pref-1 did not inhibit adipocyte differentiation of 3T3-L1 cells when differentiation was induced by standard treatment with methylisobutylxanthine, dexamethasone, and high concentrations of insulin. However, forced expression of either form of Pref-1/FA1 in 3T3-L1 or 3T3-F442A cells inhibited adipocyte differentiation when insulin or insulin-like growth factor-1 (IGF-1) was omitted from the differentiation mixture. We demonstrate that the level of the mature form of the IGF-1 receptor is reduced and that IGF-1-dependent activation of p42/p44 mitogen-activated protein kinases (MAPKs) is compromised in preadipocytes with forced expression of Pref-1. This is accompanied by suppression of clonal expansion and terminal differentiation. Accordingly, supplementation with insulin or IGF-1 rescued p42/p44 MAPK activation, clonal expansion, and adipocyte differentiation in a dose-dependent manner.
Collapse
Affiliation(s)
- Hongbin Zhang
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M., Denmark
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Krampera M, Glennie S, Dyson J, Scott D, Laylor R, Simpson E, Dazzi F. Bone marrow mesenchymal stem cells inhibit the response of naive and memory antigen-specific T cells to their cognate peptide. Blood 2003; 101:3722-9. [PMID: 12506037 DOI: 10.1182/blood-2002-07-2104] [Citation(s) in RCA: 1142] [Impact Index Per Article: 51.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have been recently shown to inhibit T-cell proliferation to polyclonal stimuli. We characterized the effect of MSCs of bone marrow origin on the T-cell response of naive and memory T cells to their cognate antigenic epitopes. The immune response to murine male transplantation antigens, HY, was selected because the peptide identity and major histocompatibility complex (MHC) restriction of the immunodominant epitopes are known. C57BL/6 female mice immunized with male cells were the source of memory T cells, whereas C6 mice transgenic for HY-specific T-cell receptor provided naive T cells. Responder cells were stimulated in vitro with male spleen cells or HY peptides in the presence or absence of MSCs. MSCs inhibited HY-specific naive and memory T cells in a dose-dependent fashion and affected cell proliferation, cytotoxicity, and the number of interferon gamma (IFN-gamma)-producing HY peptide-specific T cells. However, the MSC inhibitory effect did not selectively target antigen-reactive T cells. When MSCs were added to the T-cell cultures in a Transwell system or MSCs were replaced by MSC culture supernatant, the inhibitory activity was abrogated. T-cell reactivity was also restored if MSCs were removed from the cultures. The expression of MHC molecules and the presence in culture of antigen-presenting cells (APCs) or of CD4(+)/CD25(+) regulatory T cells were not required for MSCs to inhibit. We conclude that MSCs inhibit naive and memory T-cell responses to their cognate antigens. Overall our data suggest that MSCs physically hinder T cells from the contact with APCs in a noncognate fashion.
Collapse
Affiliation(s)
- Mauro Krampera
- Department of Immunology and Transplantation Biology, Imperial College Faculty of Medicine, Hammersmith Hospital, London, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
37
|
Castro-Muñozledo F, Beltrán-Langarica A, Kuri-Harcuch W. Commitment of 3T3-F442A cells to adipocyte differentiation takes place during the first 24-36 h after adipogenic stimulation: TNF-alpha inhibits commitment. Exp Cell Res 2003; 284:163-72. [PMID: 12651150 DOI: 10.1016/s0014-4827(02)00036-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
We studied the commitment of 3T3-F442A cells during stimulation with adipogenic serum or growth hormone. Confluent 3T3-F442A preadipocytes were incubated with adipogenic medium for increasing times; the number of adipose clusters, GPDH activity, and lipid accumulation were evaluated. Results show that cell commitment took place during the first 24-36 h after stimulation under adipogenic conditions. Then, cultures underwent a 2-fold increase in total cell number through selective multiplication of committed cells, followed by a dramatic decrease in colony-forming ability and 300- to 1000-fold raise in GPDH activity. Cell commitment was not modulated by insulin, but this hormone stimulated clonal expansion of committed cells and lipogenesis. Commitment was inhibited by TNF-alpha at concentrations as low as 5 ng/ml, and by retinoic acid. The results show that TNF-alpha inhibits adipose conversion at two different levels: at concentrations as low as 5 ng/ml, it blocks commitment, and at concentrations of 100 ng/ml or higher the cytokine seems to block mitotic expansion and other steps of differentiation after cell commitment. The identification of a specific time for cell commitment would allow the study of the early genes that might regulate cell reprogramming into adipocytes.
Collapse
Affiliation(s)
- Federico Castro-Muñozledo
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del I.P.N. Apdo. Postal 14-740, Mexico City, 07000, Mexico
| | | | | |
Collapse
|
38
|
Joudrey EM, Lechniak D, Petrik J, King WA. Expression of growth hormone and its transcription factor, Pit-1, in early bovine development. Mol Reprod Dev 2003; 64:275-83. [PMID: 12548660 DOI: 10.1002/mrd.10237] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
During bovine embryogenesis, bovine growth hormone (bGH) contributes to proliferation, differentiation, and modulation of embryo metabolism. Pituitary-specific transcription factor-1 (Pit-1) is a transcription factor that binds to promoters of GH, prolactin (PRL), and thyroid-stimulating hormone-beta (TSHbeta) encoding genes. A polymorphism in the fifth exon of the bGH gene resulting in a leucine (Leu) to valine (Val) substitution provides an Alu I restriction site when the Leu allele is present. To determine the onset of embryonic expression of the bGH gene, oocytes derived from ovaries homozygous for Leu alleles were fertilized in vitro with spermatozoa obtained from a Val homozygote. For each developmental stage examined, three separate pools of embryos composed of approximately 100 cell samples underwent RNA isolation, reverse transcription to cDNA, and amplification by nested PCR (nPCR). Bovine GH gene transcripts were identified at 2- to 4-cell (n = 162), 8- to 16-cell (n = 73), morulae (n = 51), and blastocyst (n = 15) stages. Likewise, transcripts for Pit-1 were detected at 2-cell (n = 125), 4-cell (n = 114), 8-cell (n = 56), 12-to-32-cell (n = 32), morulae (n = 68), and blastocyst (n = 14) stages. After digestion with Alu1, bGH cDNA was genotyped by restriction fragment length polymorphism (RFLP) analysis. Bovine GH mRNA was present in all pools of stages examined. Both Leu and Val alleles (maternal and paternal) were only detected in pools of embryos that had reached 8- to 16-cell stage. Results suggest that transcription of the bGH gene begins at the 8- to 16-cell stage in bovine embryos, possibly under control of the transcription factor, Pit-1, and that RFLP analysis of the bGH gene can be used to determine parental origin of transcripts in early embryonic development.
Collapse
Affiliation(s)
- E M Joudrey
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | | | | | | |
Collapse
|
39
|
Kopchick JJ, Parkinson C, Stevens EC, Trainer PJ. Growth hormone receptor antagonists: discovery, development, and use in patients with acromegaly. Endocr Rev 2002; 23:623-46. [PMID: 12372843 DOI: 10.1210/er.2001-0022] [Citation(s) in RCA: 210] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
An understanding of the events that occur during GH receptor (GHR) signaling has facilitated the development of a GHR antagonist (pegvisomant) for use in humans. This molecule has been designed to compete with native GH for the GHR and to prevent its proper or functional dimerization-a process that is critical for GH signal transduction and IGF-I synthesis and secretion. Clinical trials in patients with acromegaly show GHR blockade to be an exciting new mode of therapy for this condition, and pegvisomant may have a therapeutic role in diseases, such as diabetes and malignancy, in which abnormalities of the GH/IGF-I axis have been observed. This review charts the discovery and development of GHR antagonists and details the experience gained in patients with acromegaly.
Collapse
Affiliation(s)
- J J Kopchick
- Edison Biotechnology Institute, Department of Biomedical Sciences, College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701, USA
| | | | | | | |
Collapse
|
40
|
Greenhalgh CJ, Bertolino P, Asa SL, Metcalf D, Corbin JE, Adams TE, Davey HW, Nicola NA, Hilton DJ, Alexander WS. Growth enhancement in suppressor of cytokine signaling 2 (SOCS-2)-deficient mice is dependent on signal transducer and activator of transcription 5b (STAT5b). Mol Endocrinol 2002; 16:1394-406. [PMID: 12040024 DOI: 10.1210/mend.16.6.0845] [Citation(s) in RCA: 104] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Mice lacking suppressor of cytokine signaling-2 (SOCS-2) exhibit accelerated postnatal growth resulting in adult mice that are 1.3 to 1.5 times the size of normal mice. In this study we examined the somatotrophic pathway to determine whether the production or actions of GH or IGF-I are altered in these mice. We demonstrated that SOCS-2(-/-) mice do not have elevated GH levels and suffer no major pituitary dysmorphogenesis, and that SOCS-2-deficient embryonic fibroblasts do not have altered IGF-I signaling. Primary hepatocytes from SOCS-2(-/-) mice, however, did have moderately prolonged signal transducer and activator of transcription 5 signaling in response to GH stimulation. Furthermore, the deletion of SOCS-2 from mice also lacking signal transducer and activator of transcription 5b had little effect on growth, suggesting that the action of SOCS-2 may be the regulation of the GH signaling pathway.
Collapse
Affiliation(s)
- Christopher J Greenhalgh
- The Cooperative Research Centre for Cellular Growth Factors and the Walter and Eliza Hall Institute of Medical Research, Royal Melbourne Hospital, Victoria, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Shang CA, Thompson BJL, Teasdale R, Brown RJ, Waters MJ. Genes induced by growth hormone in a model of adipogenic differentiation. Mol Cell Endocrinol 2002; 189:213-9. [PMID: 12039079 DOI: 10.1016/s0303-7207(01)00590-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
A substantial number of GH regulated genes have been reported in mature hepatocytes, but genes involved in GH-initiated cell differentiation have not yet been identified. Here we have studied a well-characterised model of GH-dependent differentiation, adipogenesis of 3T3-F442A preadipocytes, to identify genes rapidly induced by GH. Using the suppression subtractive hybridisation technique, we have identified eight genes induced within 60 min of GH treatment, and verified these by northern analysis. Six were identifiable as Stat 2, Stat 3, thrombospondin-1, oncostatin M receptor beta chain, a DEAD box RNA helicase, and muscleblind, a developmental transcription factor. Bioinformatic approaches assigned one of the two remaining unknown genes as a novel 436 residue serine/threonine kinase. As each of the identified genes have important developmental roles, they may be important in initiating GH-induced adipogenesis.
Collapse
Affiliation(s)
- Catherine A Shang
- Department of Physiology and Pharmacology, Institute for Molecular Biology, University of Queensland, Qld 4072, Brisbane, Australia
| | | | | | | | | |
Collapse
|
42
|
Jeay S, Sonenshein GE, Postel-Vinay MC, Kelly PA, Baixeras E. Growth hormone can act as a cytokine controlling survival and proliferation of immune cells: new insights into signaling pathways. Mol Cell Endocrinol 2002; 188:1-7. [PMID: 11911939 DOI: 10.1016/s0303-7207(02)00014-x] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
While growth hormone (GH) is classically defined as a peptide hormone, recent evidence supports a role for GH acting as a cytokine in the immune system under conditions of stress, counteracting immunosuppression by glucocorticoids. Lymphoid cells express the GH receptor, which belongs to the cytokine receptor superfamily, and GH can be produced by immune tissues, suggesting an autocrine/paracrine mode of action of GH. GH can act as a cytokine, promoting cell cycle progression of lymphoid cells and preventing apoptosis. These effects of GH were shown to be mainly mediated by the PI-3 kinase/Akt pathway and the transcription factor NF-kappaB. Expression of several cell cycle mediators, as well as Bcl-2, c-Myc and cyclin proteins were found to be regulated by GH. Survival of immune cells under conditions of stress was promoted by NF-kappaB. Thus, GH acts not only as a hormone but also as a cytokine, playing a potentially important role in immune system cells. Lastly, in this mini-review, we will discuss whether the discovery of these molecules in GH signaling pathways offers new insights into additional mechanisms of action whereby GH regulates apoptosis, proliferation and neoplastic transformation of cells of the immune system.
Collapse
Affiliation(s)
- Sébastien Jeay
- INSERM Unité 344, Endocrinologie Moléculaire, Faculté de Médecine Necker, 75730 Paris Cedex 15, France.
| | | | | | | | | |
Collapse
|
43
|
|
44
|
Butler AA, Le Roith D. Control of growth by the somatropic axis: growth hormone and the insulin-like growth factors have related and independent roles. Annu Rev Physiol 2001; 63:141-64. [PMID: 11181952 DOI: 10.1146/annurev.physiol.63.1.141] [Citation(s) in RCA: 239] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The traditionally accepted theory has been that most of the biological effects of growth hormone (GH) are mediated by circulating (endocrine) insulin-like growth factor-I (IGF-I). This dogma was modified when it was discovered that most tissues express IGF-I that can act via an autocrine/paracrine fashion. In addition, both GH and IGF-I had independent effects on various target tissues. Using tissue-specific gene deletion of IGF-I in the liver, it has been shown that circulating IGF-I is predominantly liver-derived but is not essential for normal postnatal growth. Therefore, it is proposed that non-hepatic tissue-derived IGF-I may be sufficient for growth and development. Thus the original somatomedin hypothesis has undergone further modifications.
Collapse
Affiliation(s)
- A A Butler
- The Vollum Institute for Advanced Biomedical Research, Oregon Health Sciences University, Portland Oregon 97201-3098, USA.
| | | |
Collapse
|
45
|
Abstract
Since the original somatomedin hypothesis was conceived, a number of important discoveries have allowed investigators to modify the concept. Originally somatic growth was thought to be controlled by pituitary GH and mediated by circulating insulin-like growth factor-I (IGF-I, somatomedin C) expressed exclusively by the liver. With the discovery that IGF-I is produced by most, if not all, tissues, the role of autocrine/paracrine IGF-I vs. the circulating form has been hotly debated. Recent experiments using transgenic and gene-deletion technologies have attempted to answer these questions. In the liverspecific igf-1 gene-deleted mouse model, postnatal growth and development are normal despite the marked reduction in circulating IGF-I and IGF-binding protein levels; free IGF-I levels are normal. Thus, the normal postnatal growth and development in these animals may be due to normal free IGF-I levels (from as yet unidentified sources), although the role of autocrine/paracrine IGF-I has yet to be determined.
Collapse
Affiliation(s)
- D Le Roith
- Clinical Endocrinology Branch, National Institutes of Health, Bethesda, Maryland 20892-1758, USA.
| | | | | | | | | |
Collapse
|
46
|
Abstract
Growth hormone (GH) is not only the major regulator of postnatal somatic growth but also exerts profound effects on body composition through a combination of anabolic, lipolytic and antinatriuretic actions. GH enhancement of the lipolytic activity of adipose tissue in combination with a reduction of triglyceride accumulation via inhibition of lipoprotein lipase activity appears to be the major mechanism by which GH results in a reduction of the total fat mass. Recently, much progress has been made in understanding the molecular mechanism by which GH affects cellular function. This review provides a brief discourse and summary of the mechanism of effects of GH on preadipocyte/adipocyte function. It is intended to provide a functional understanding of the mechanism of action of GH as it relates to adipogenesis and adipocyte function.
Collapse
Affiliation(s)
- S Y Nam
- Department of Internal Medicine, Yongdong Severance Hospital, Yonsei University College of Medicine, Dogok-dong 146-92, Kangnam-Ku, Seoul 135-270, Korea
| | | |
Collapse
|
47
|
Affiliation(s)
- J J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701, USA
| | | |
Collapse
|
48
|
Ohlsson C, Lindahl A, Isgaard J, Nilsson A, Isaksson OGP. The Dual Effector Theory. Compr Physiol 1999. [DOI: 10.1002/cphy.cp070516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
49
|
Juárez-Aguilar E, Castro-Muñozledo F, Guerra-Rodríguez NE, Reséndez-Pérez D, Martínez-Rodríguez HG, Barrera-Saldaña HA, Kuri-Harcuch W. Functional domains of human growth hormone necessary for the adipogenic activity of hGH/hPL chimeric molecules. J Cell Sci 1999; 112 Pt 18:3127-35. [PMID: 10462528 DOI: 10.1242/jcs.112.18.3127] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Genetic analysis through construction of chimeric genes and their transfection in mammalian cells could provide a better understanding of biological functions of native or modified proteins, and would allow the design of new gene constructs encoding peptides that mimic or block ligand interaction with target tissues. To identify the hGH domains responsible for induction of adipose differentiation we constructed hGH/hPL chimeric molecules using homologous DNA mutagenesis, since hGH, but not human placental lactogen (hPL), promotes adipose differentiation in mouse 3T3-F442A cells. We assayed their adipogenic activity in an autocrine/paracrine biological model consisting of transiently transfected 3T3-F442A cells with the chimeric constructs. Plasmid DNAs carrying these constructs were transfected into growing 3T3-F442A cells, and cultures were further maintained for 7 days to differentiate into adipocytes. Secretion of transfected hGH/hPL chimeric proteins into the medium was in the range of 5–25 ng/ml. Adipogenic activity was a property only of those chimeric proteins that contained hGH exon III together with either hGH exon II or hGH IV. Our results also suggest that hGH binding site-2 is composed of two structural subdomains: subsite 2A encoded by exon II of hGH and subsite-2B encoded by exon IV. We also suggest that full adipogenic activity requires the presence of binding site-1 and any of the subsites of binding site-2. This simple autocrine/paracrine biological model of gene transfection allows the analysis of specific biological activity of products encoded by modified genes.
Collapse
Affiliation(s)
- E Juárez-Aguilar
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del I.P.N., Apdo. Postal 14-740 Mexico, D.F. 07000, Mexico
| | | | | | | | | | | | | |
Collapse
|
50
|
Gerfault V, Louveau I, Mourot J. The effect of GH and IGF-I on preadipocytes from Large White and Meishan pigs in primary culture. Gen Comp Endocrinol 1999; 114:396-404. [PMID: 10336827 DOI: 10.1006/gcen.1999.7271] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Proliferation and differentiation of preadipocytes from 7-day-old Large White (LW) and Meishan (MS) pigs were studied in primary culture. The effects of porcine GH (pGH) and IGF-I as well as the expression of GH (GHR) and IGF-I (IGF-IR) receptors mRNA were examined. Preadipocytes were exposed to serum-supplemented and serum-free medium to determine proliferation and differentiation, respectively. Proliferation was higher in MS than in LW pigs. Treatment with pGH (2 nM) or IGF-I (10 nM) resulted in a similar decrease in proliferation in LW and MS pigs. Parameters assessing differentiation and the effects of pGH and IGF-I on differentiation did not differ between the two breeds. The percentage of differentiating cells and LPL and ME activities were markedly reduced by pGH. IGF-I did not reduce differentiation significantly. Both GHR and IGF-IR mRNA were expressed in adipose tissue, adipocytes, preadipocytes, and 6-day-cultured cells from LW and MS pigs. The similar action of pGH and IGF-I on preadipocyte proliferation and differentiation, associated with the similar expression of GHR and IGF-IR mRNA in LW and MS pigs, suggests that the GH/IGF-I axis is not impaired in MS pigs. The difference in preadipocyte proliferation observed between LW and MS pigs could account for their adiposity difference.
Collapse
Affiliation(s)
- V Gerfault
- Station de Recherches Porcines, Saint Gilles, 35590, France
| | | | | |
Collapse
|