401
|
Venken K, Movérare-Skrtic S, Kopchick JJ, Coschigano KT, Ohlsson C, Boonen S, Bouillon R, Vanderschueren D. Impact of androgens, growth hormone, and IGF-I on bone and muscle in male mice during puberty. J Bone Miner Res 2007; 22:72-82. [PMID: 17014385 DOI: 10.1359/jbmr.060911] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
UNLABELLED The interaction between androgens and GH/IGF-I was studied in male GHR gene disrupted or GHRKO and WT mice during puberty. Androgens stimulate trabecular and cortical bone modeling and increase muscle mass even in the absence of a functional GHR. GHR activation seems to be the main determinant of radial bone expansion, although GH and androgens are both necessary for optimal stimulation of periosteal growth during puberty. INTRODUCTION Growth hormone (GH) is considered to be a major regulator of postnatal skeletal growth, whereas androgens are considered to be a key regulator of male periosteal bone expansion. Moreover, both androgens and GH are essential for the increase in muscle mass during male puberty. Deficiency or resistance to either GH or androgens impairs bone modeling and decreases muscle mass. The aim of the study was to investigate androgen action on bone and muscle during puberty in the presence and absence of a functional GH/insulin-like growth factor (IGF)-I axis. MATERIALS AND METHODS Dihydrotestosterone (DHT) or testosterone (T) were administered to orchidectomized (ORX) male GH receptor gene knockout (GHRKO) and corresponding wildtype (WT) mice during late puberty (6-10 weeks of age). Trabecular and cortical bone modeling, cortical strength, body composition, IGF-I in serum, and its expression in liver, muscle, and bone were studied by histomorphometry, pQCT, DXA, radioimmunoassay and RT-PCR, respectively. RESULTS GH receptor (GHR) inactivation and low serum IGF-I did not affect trabecular bone modeling, because trabecular BMD, bone volume, number, width, and bone turnover were similar in GHRKO and WT mice. The normal trabecular phenotype in GHRKO mice was paralleled by a normal expression of skeletal IGF-I mRNA. ORX decreased trabecular bone volume significantly and to a similar extent in GHRKO and WT mice, whereas DHT and T administration fully prevented trabecular bone loss. Moreover, DHT and T stimulated periosteal bone formation, not only in WT (+100% and +100%, respectively, versus ORX + vehicle [V]; p < 0.05), but also in GHRKO mice (+58% and +89%, respectively, versus ORX + V; p < 0.05), initially characterized by very low periosteal growth. This stimulatory action on periosteal bone resulted in an increase in cortical thickness and occurred without any treatment effect on serum IGF-I or skeletal IGF-I expression. GHRKO mice also had reduced lean body mass and quadriceps muscle weight, along with significantly decreased IGF-I mRNA expression in quadriceps muscle. DHT and T equally stimulated muscle mass in GHRKO and WT mice, without any effect on muscle IGF-I expression. CONCLUSIONS Androgens stimulate trabecular and cortical bone modeling and increase muscle weight independently from either systemic or local IGF-I production. GHR activation seems to be the main determinant of radial bone expansion, although GHR signaling and androgens are both necessary for optimal stimulation of periosteal growth during puberty.
Collapse
|
402
|
Choi JH, Kim HS, Kim SH, Yang YR, Bae YS, Chang JS, Kwon HM, Ryu SH, Suh PG. Phospholipase Cgamma1 negatively regulates growth hormone signalling by forming a ternary complex with Jak2 and protein tyrosine phosphatase-1B. Nat Cell Biol 2006; 8:1389-97. [PMID: 17128263 DOI: 10.1038/ncb1509] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2006] [Accepted: 10/06/2006] [Indexed: 01/20/2023]
Abstract
Growth hormone binds to its membrane receptor (GHR), whereby it regulates many cellular functions, including proliferation, differentiation and chemotaxis. However, although the activation of growth hormone-mediated signalling is well understood, the precise mechanism responsible for its regulation has not been elucidated. Here, we demonstrate that phospholipase Cgamma1 (PLCgamma1) modulates the action of growth hormone-mediated signalling by interacting with tyrosine kinase Jak2 (janus kinase 2) in a growth hormone-dependent manner. In the absence of PLCgamma1 (PLCgamma1(-/-)), growth hormone-induced JAK2 and STAT5 phosphorylation significantly increased in mouse embryonic fibroblasts (MEFs). Furthermore, the re-expression of PLCgamma1 reduced growth hormone-induced Jak2 activation. Growth hormone-induced Jak2 phosphorylation was enhanced by siRNA-specific knockdown of PLCgamma1. Interestingly, PLCgamma1 physically linked Jak2 and protein tyrosine phosphatase-1B (PTP-1B) by binding to both using different domains, and this process was implicated in the modulation of cytokine signalling through Jak2. In addition, in PLCgamma1(-/-) MEFs, growth hormone-dependent c-Fos activation was upregulated and growth hormone-induced proliferation was potentiated. These results suggest that PLCgamma1 has a key function in the regulation of growth hormone-mediated signalling by negatively regulating Jak2 activation.
Collapse
Affiliation(s)
- Jang Hyun Choi
- National Research Laboratory of Signaling Network, Department of Life Science, Pohang University of Science and Technology, Pohang, Kyungbuk, 790-784, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
403
|
Abstract
Sustaining health and extending longevity have been perpetual goals of all human societies. For almost as long, there has been an ongoing effort to develop treatments that could prevent aging and, more importantly, make us live longer and more healthily. At present, there is one known intervention that delays aging, increases lifespan and prevents diseases in many animal species: calorie restriction. There are other physiological factors that are believed to have corresponding impacts on longevity and aging, including growth hormone and the insulin/insulin-like growth factor 1 signaling pathway. However, there is still much debate regarding the complex action of growth hormone on lifespan and aging.
Collapse
Affiliation(s)
- Michal M Masternak
- a Southern Illinois University, School of Medicine, Geriatrics Research, Department of Internal Medicine, 801 N. Rutledge Street, Room 4389, PO Box 19628, Springfield, IL 62794-9628, USA.
| | - Andrzej Bartke
- b Southern Illinois University, School of Medicine, Geriatrics Research, Department of Internal Medicine and Physiology, 801 N. Rutledge Street, Room 4389, PO Box 19628, Springfield, IL 62794-9628, USA.
| |
Collapse
|
404
|
Tshori S, Gilon D, Beeri R, Nechushtan H, Kaluzhny D, Pikarsky E, Razin E. Transcription factor MITF regulates cardiac growth and hypertrophy. J Clin Invest 2006; 116:2673-81. [PMID: 16998588 PMCID: PMC1570375 DOI: 10.1172/jci27643] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2005] [Accepted: 08/08/2006] [Indexed: 12/19/2022] Open
Abstract
High levels of microphthalmia transcription factor (MITF) expression have been described in several cell types, including melanocytes, mast cells, and osteoclasts. MITF plays a pivotal role in the regulation of specific genes in these cells. Although its mRNA has been found to be present in relatively high levels in the heart, its cardiac role has never been explored. Here we show that a specific heart isoform of MITF is expressed in cardiomyocytes and can be induced by beta-adrenergic stimulation but not by paired box gene 3 (PAX3), the regulator of the melanocyte MITF isoform. In 2 mouse strains with different MITF mutations, heart weight/body weight ratio was decreased as was the hypertrophic response to beta-adrenergic stimulation. These mice also demonstrated a tendency to sudden death following beta-adrenergic stimulation. Most impressively, 15-month-old MITF-mutated mice had greatly decreased heart weight/body weight ratio, systolic function, and cardiac output. In contrast with normal mice, in the MITF-mutated mice, beta-adrenergic stimulation failed to induce B-type natriuretic peptide (BNP), an important modulator of cardiac hypertrophy, while atrial natriuretic peptide levels and phosphorylated Akt were increased, suggesting a cardiac stress response. In addition, cardiomyocytes cultured with siRNA against MITF showed a substantial decrease in BNP promoter activity. Thus, for what we believe is the first time, we have demonstrated that MITF plays an essential role in beta-adrenergic-induced cardiac hypertrophy.
Collapse
Affiliation(s)
- Sagi Tshori
- Department of Biochemistry, Hebrew University Medical School, Jerusalem, Israel.
Heart Institute,
Department of Oncology, and
Department of Pathology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Dan Gilon
- Department of Biochemistry, Hebrew University Medical School, Jerusalem, Israel.
Heart Institute,
Department of Oncology, and
Department of Pathology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Ronen Beeri
- Department of Biochemistry, Hebrew University Medical School, Jerusalem, Israel.
Heart Institute,
Department of Oncology, and
Department of Pathology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Hovav Nechushtan
- Department of Biochemistry, Hebrew University Medical School, Jerusalem, Israel.
Heart Institute,
Department of Oncology, and
Department of Pathology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Dmitry Kaluzhny
- Department of Biochemistry, Hebrew University Medical School, Jerusalem, Israel.
Heart Institute,
Department of Oncology, and
Department of Pathology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Eli Pikarsky
- Department of Biochemistry, Hebrew University Medical School, Jerusalem, Israel.
Heart Institute,
Department of Oncology, and
Department of Pathology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Ehud Razin
- Department of Biochemistry, Hebrew University Medical School, Jerusalem, Israel.
Heart Institute,
Department of Oncology, and
Department of Pathology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
405
|
Growth hormone and insulin-like growth factor-I act together and independently when regulating growth in vertebral and muscle tissue of atlantic salmon postsmolts. Gen Comp Endocrinol 2006; 149:253-60. [PMID: 16890227 DOI: 10.1016/j.ygcen.2006.06.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2005] [Revised: 05/30/2006] [Accepted: 06/08/2006] [Indexed: 02/02/2023]
Abstract
Aiming to elucidate the role of GH and IGF-I with regard to vertebral and white muscle growth, gene expression of the GH and IGF-I receptors (ghr and igf-Ir, respectively) and local IGF-I (igf-I) were analyzed during spring growth (January-June) in Atlantic salmon postsmolts. One group of fish was reared under natural light (NL), while one group was reared under continuous light (LL). Growth rate of fork length was higher in the LL group for a short period after onset of continuous light (LL: 0.50+/-0.02 mm day(-1), NL: 0.43+/-0.01 mm day(-1)) and for a longer period at the end of the experiment in June (LL: 1.18+/-0.06 mm day(-1), NL: 0.75+/-0.02 mm day(-1)). Likewise, growth rate in length of vertebra No. 40 in the LL group was higher than in the NL group the first period after onset of light (LL: 0.015+/-0.002 mm day(-1), NL: 0.008+/-0.001 mm day(-1)). Plasma GH levels peaked in late February and were higher in the LL group than in the NL group (LL: 7.27+/-0.61 ng ml(-1), NL: 2.60+/-0.50 ng ml(-1)), whereas plasma IGF-I levels peaked in early February and were unaffected by photoperiod. ghr expression was upregulated in late February in liver (12-fold), white muscle (6-fold) and vertebral tissue (3-fold) and higher in the LL group than in the NL group (2-fold) in vertebral tissue in late March. White muscle expression of igf-I and igf-Ir decreased from initial levels throughout the experiment. Hepatic gene expression of igf-I doubled in both groups in late February, followed by a 4-fold upregulation in June in the LL group only. Vertebral tissue expression of igf-I (4-fold) and igf-Ir (6-fold) increased in May and were unaffected by photoperiod. One exception was a smaller upregulation of igf-I (2-fold) in the LL group in early February. In conclusion, GH appears to have an initial role in stimulating vertebral growth, while IGF-I seems to stimulate growth during late spring. It is suggested that local IGF-I acts as a paracrine agent, evaluated from the concurrent upregulation of igf-I and igf-Ir. The upregulation of ghr in white muscle tissue, concurrent with a downregulation of muscle igf-I and igf-Ir, indicate that GH stimulated growth or metabolism independent of IGF-I.
Collapse
|
406
|
Delahunty KM, Shultz KL, Gronowicz GA, Koczon-Jaremko B, Adamo ML, Horton LG, Lorenzo J, Donahue LR, Ackert-Bicknell C, Kream BE, Beamer WG, Rosen CJ. Congenic mice provide in vivo evidence for a genetic locus that modulates serum insulin-like growth factor-I and bone acquisition. Endocrinology 2006; 147:3915-23. [PMID: 16675518 DOI: 10.1210/en.2006-0277] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
We identified quantitative trait loci (QTL) that determined the genetic variance in serum IGF-I through genome-wide scanning of mice derived from C57BL/6J(B6) x C3H/HeJ(C3H) intercrosses. One QTL (Igf1s2), on mouse chromosome 10 (Chr10), produces a 15% increase in serum IGF-I in B6C3 F2 mice carrying c3 alleles at that position. We constructed a congenic mouse, B6.C3H-10 (10T), by backcrossing c3 alleles from this 57-Mb region into B6 for 10 generations. 10T mice have higher serum and skeletal IGF-I, greater trabecular bone volume fraction, more trabeculae, and a higher number of osteoclasts at 16 wk, compared with B6 (P < 0.05). Nested congenic sublines generated from further backcrossing of 10T allowed for recombination and produced four smaller sublines with significantly increased serum IGF-I at 16 wk (i.e. 10-4, 10-7, 10-10, and 10-13), compared with B6 (P < 0.0003), and three smaller sublines that showed no differences in IGF-I vs. age- and gender-matched B6 mice. Like 10T, the 10-4 nested sublines at 16 wk had higher femoral mineral (P < 0.0001) and greater trabecular connectivity density with significantly more trabeculae than B6 (P < 0.01). Thus, by comprehensive phenotyping, we were able to narrow the QTL to an 18.3-Mb region containing approximately 148 genes, including Igf1 and Elk-3(ETS domain protein). Allelic differences in the Igf1s2 QTL produce a phenotype characterized by increased serum IGF-I and greater peak bone density. Congenic mice establish proof of concept of shared genetic determinants for both circulating IGF-I and bone acquisition.
Collapse
Affiliation(s)
- K M Delahunty
- The Jackson Laboratory, 600 Main Street, Bar Harbor, Maine 04609, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
407
|
Chen X, Shevtsov SP, Hsich E, Cui L, Haq S, Aronovitz M, Kerkelä R, Molkentin JD, Liao R, Salomon RN, Patten R, Force T. The beta-catenin/T-cell factor/lymphocyte enhancer factor signaling pathway is required for normal and stress-induced cardiac hypertrophy. Mol Cell Biol 2006; 26:4462-73. [PMID: 16738313 PMCID: PMC1489123 DOI: 10.1128/mcb.02157-05] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
In cells capable of entering the cell cycle, including cancer cells, beta-catenin has been termed a master switch, driving proliferation over differentiation. However, its role as a transcriptional activator in terminally differentiated cells is relatively unknown. Herein we utilize conditional, cardiac-specific deletion of the beta-catenin gene and cardiac-specific expression of a dominant inhibitory mutant of Lef-1 (Lef-1Delta20), one of the members of the T-cell factor/lymphocyte enhancer factor (Tcf/Lef) family of transcription factors that functions as a coactivator with beta-catenin, to demonstrate that beta-catenin/Tcf/Lef-dependent gene expression regulates both physiologic and pathological growth (hypertrophy) of the heart. Indeed, the profound nature of the growth impairment of the heart in the Lef-1Delta20 mouse, which leads to very early development of heart failure and premature death, suggests beta-catenin/Tcf/Lef targets are dominant regulators of cardiomyocyte growth. Thus, our studies, employing complementary models in vivo, implicate beta-catenin/Tcf/Lef signaling as an essential growth-regulatory pathway in terminally differentiated cells.
Collapse
Affiliation(s)
- Xin Chen
- Molecular Cardiology Research Institute, Tufts-New England Medical Center and Tufts University School of Medicine, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
408
|
Long F, Joeng KS, Xuan S, Efstratiadis A, McMahon AP. Independent regulation of skeletal growth by Ihh and IGF signaling. Dev Biol 2006; 298:327-33. [PMID: 16905129 DOI: 10.1016/j.ydbio.2006.06.042] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2006] [Revised: 06/20/2006] [Accepted: 06/26/2006] [Indexed: 12/30/2022]
Abstract
The insulin-like growth factors (IGFs) play a major role in regulating the systemic growth of mammals. However, it is unclear to what extent their systemic and/or local functions act in concert with other local growth factors controlling the sizes of individual organs. We have specifically addressed whether growth control of the skeleton by IGFs interacts genetically with that by Indian hedgehog (Ihh), a locally produced growth signal for the endochondral skeleton. Here, we report that disruption of both IGF and Ihh signaling resulted in additive reduction in the size of the embryonic skeleton. Thus, IGF and Ihh signaling appear to control the growth of the skeleton in parallel pathways.
Collapse
Affiliation(s)
- Fanxin Long
- Department of Medicine, Washington University Medical School, St. Louis, MO 63110, USA.
| | | | | | | | | |
Collapse
|
409
|
Shavlakadze T, Boswell JM, Burt DW, Asante EA, Tomas FM, Davies MJ, White JD, Grounds MD, Goddard C. Rskalpha-actin/hIGF-1 transgenic mice with increased IGF-I in skeletal muscle and blood: impact on regeneration, denervation and muscular dystrophy. Growth Horm IGF Res 2006; 16:157-73. [PMID: 16716629 DOI: 10.1016/j.ghir.2005.11.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2005] [Revised: 11/08/2005] [Accepted: 11/09/2005] [Indexed: 10/24/2022]
Abstract
Human IGF-I was over-expressed in skeletal muscles of C57/BL6xCBA mice under the control of the rat skeletal alpha-actin gene promoter. RT-PCR verified expression of the transgene in skeletal muscle but not in the liver of 1- and 21-day old heterozygote transgenic mice. The concentration of endogenous mouse IGF-I, measured by an immunoassay which does not detect human IGF-I, was not significantly different between transgenic mice and wild-type littermates (9.5 +/- 0.8 and 13.3 +/- 1.9 ng/g in muscle; 158.3 +/- 18.6 and 132.9 +/- 33.1 ng/ml in plasma, respectively). In contrast, quantitation with antibodies to human IGF-I showed an increase in IGF-I of about 100 ng/ml in plasma and 150 ng/g in muscle of transgenic mice at 6 months of age. Transgenic males, compared to their age matched wild-type littermates, had a significantly higher body weight (38.6 +/- 0.53 g vs. 35.8 +/- 0.64 g at 6 months of age; P < 0.001), dry fat-free carcass mass (5.51 +/- 0.085 vs. 5.08 +/- 0.092 g; P < 0.001) and myofibrillar protein mass (1.62 +/- 0.045 vs. 1.49 +/- 0.048 g; P < 0.05), although the fractional content of fat in the carcass was lower (167 +/- 7.0 vs. 197 +/- 7.7 g/kg wet weight) in transgenic animals. There was no evidence of muscle hypertrophy and no change in the proportion of slow type I myofibres in the limb muscles of Rskalpha-actin/hIGF-I transgenic mice at 3 or 6 months of age. Phenotypic changes in Rskalpha-actin/hIGF-I mice are likely to be due to systemic as well as autocrine/paracrine effects of overproduction of IGF-I due to expression of the human IGF-I transgene. The effect of muscle specific over-expression of Rskalpha-actin/hIGF-I transgene was tested on: (i) muscle regeneration in auto-transplanted whole muscle grafts; (ii) myofibre atrophy following sciatic nerve transection; and (iii) sarolemmal damage and myofibre necrosis in dystrophic mdx muscle. No beneficial effect of muscle specific over-expression of Rskalpha-actin/hIGF-I transgene was seen in these three experimental models.
Collapse
Affiliation(s)
- T Shavlakadze
- School of Anatomy and Human Biology, The University of Western Australia, Crawley, Perth.
| | | | | | | | | | | | | | | | | |
Collapse
|
410
|
He J, Rosen CJ, Adams DJ, Kream BE. Postnatal growth and bone mass in mice with IGF-I haploinsufficiency. Bone 2006; 38:826-35. [PMID: 16427371 DOI: 10.1016/j.bone.2005.11.021] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2005] [Revised: 10/25/2005] [Accepted: 11/17/2005] [Indexed: 11/23/2022]
Abstract
We examined the influence of IGF-I haploinsufficiency on growth, bone mass and osteoblast differentiation in Igf1 heterozygous knockout (HET) mice. Cohorts of male and female wild type (WT) and HET mice in the outbred CD-1 background were analyzed at 1, 2, 4, 8, 12, 15 and 18 months of age for body weight, serum IGF-I and bone morphometry. Compared to WT mice, HET mice had 20-30% lower serum IGF-I levels in both genders and in all age groups. Female HET mice showed significant reductions in body weight (10-20%), femur length (4-6%) and femoral bone mineral density (BMD) (7-12%) before 15 months of age. Male HET mice showed significant differences in all parameters at 2 months and thereafter. At 8 and 12 months, WT mice also showed a significant gender effect: despite their lower body weight, female mice had higher femoral BMD and femur length compared to males. Microcomputed tomography showed a significant reduction in cortical bone area (7-20%) and periosteal circumference (5-13%) with no consistent pattern of change in trabecular bone measurements in 2- and 8-month old HET mice in both genders. HET primary osteoblast cultures showed a 40% reduction in IGF-I protein expression and a 50% decrease in IGF-I mRNA expression. Cell growth and proliferation were decreased in HET cultures. Thus, IGF-I haploinsufficiency in outbred male and female mice resulted in reduced body weight, femur length and areal BMD at most ages. Serum IGF-I levels showed a high level of positive correlation with body weight and skeletal morphometry. These studies show that IGF-I is a determinant of bone size and mass in postnatal life. We speculate that impaired osteoblast proliferation may contribute to the skeletal phenotype of mice with IGF-I haploinsufficiency.
Collapse
Affiliation(s)
- Jianing He
- Department of Medicine, The University of Connecticut Health Center, Farmington, 06030-1850, USA
| | | | | | | |
Collapse
|
411
|
Coschigano KT. Aging-related characteristics of growth hormone receptor/binding protein gene-disrupted mice. AGE (DORDRECHT, NETHERLANDS) 2006; 28:191-200. [PMID: 19943140 PMCID: PMC2464722 DOI: 10.1007/s11357-006-9004-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2005] [Accepted: 12/01/2005] [Indexed: 05/28/2023]
Abstract
Since generation of the growth hormone receptor/binding protein (GHR/BP) gene-disrupted mouse nearly 10 years ago, use of this mouse model has become widespread in the elucidation of the physiological roles of GH and insulin-like growth factor-1 (IGF-1). In particular, it serves as a useful model to study mechanisms of aging. This review highlights the evidence demonstrating that the loss of GH signaling leads to lifespan extension in mice, and presents the multiple characteristics of this mouse line that suggest the life extension is due to alteration of the aging process.
Collapse
Affiliation(s)
- Karen T Coschigano
- Department of Biomedical Sciences, College of Osteopathic Medicine, Ohio University, 351 Irvine Hall, Athens, OH 45701, USA.
| |
Collapse
|
412
|
Sotiropoulos A, Ohanna M, Kedzia C, Menon RK, Kopchick JJ, Kelly PA, Pende M. Growth hormone promotes skeletal muscle cell fusion independent of insulin-like growth factor 1 up-regulation. Proc Natl Acad Sci U S A 2006; 103:7315-20. [PMID: 16670201 PMCID: PMC1456062 DOI: 10.1073/pnas.0510033103] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Growth hormone (GH) participates in the postnatal regulation of skeletal muscle growth, although the mechanism of action is unclear. Here we show that the mass of skeletal muscles lacking GH receptors is reduced because of a decrease in myofiber size with normal myofiber number. GH signaling controls the size of the differentiated myotubes in a cell-autonomous manner while having no effect on size, proliferation, and differentiation of the myoblast precursor cells. The GH hypertrophic action leads to an increased myonuclear number, indicating that GH facilitates fusion of myoblasts with nascent myotubes. NFATc2, a transcription factor regulating this phase of fusion, is required for GH action because GH is unable to induce hypertrophy of NFATc2-/- myotubes. Finally, we provide three lines of evidence suggesting that GH facilitates cell fusion independent of insulin-like growth factor 1 (IGF-1) up-regulation. First, GH does not regulate IGF-1 expression in myotubes; second, GH action is not mediated by a secreted factor in conditioned medium; third, GH and IGF-1 hypertrophic effects are additive and rely on different signaling pathways. Taken together, these data unravel a specific function of GH in the control of cell fusion, an essential process for muscle growth.
Collapse
Affiliation(s)
- Athanassia Sotiropoulos
- Institut National de la Santé et de la Recherche Médicale, U810, and Faculté de Médecine Necker-Enfants Malades, Université Paris 5, F-75730 Paris, France.
| | | | | | | | | | | | | |
Collapse
|
413
|
Woodhouse LJ, Mukherjee A, Shalet SM, Ezzat S. The influence of growth hormone status on physical impairments, functional limitations, and health-related quality of life in adults. Endocr Rev 2006; 27:287-317. [PMID: 16543384 DOI: 10.1210/er.2004-0022] [Citation(s) in RCA: 126] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The availability of recombinant human GH and somatostatin analogs has resulted in widespread treatment for adults with GH deficiency (GHD) and those with GH excess (acromegaly). Despite being at opposite ends of the spectrum in terms of their GH/IGF-I axis, both of these populations experience overlapping somatic impairments. Adults with untreated GHD have low circulating levels of IGF-I that manifest as altered body composition with increased fat and reduced lean body and skeletal muscle mass. At the other end of the spectrum, adults with GH excess, who have elevated levels of IGF-I, also have altered body composition. Impairments that result from disorders of either GHD or GH excess are both associated with increased functional limitations, such as reduced ability to walk quickly for prolonged periods, and poorer health-related quality of life (HR-QoL). Adults with untreated GHD and GH excess both commonly complain of excessive fatigue that seems to be associated more with impaired aerobic than muscular performance. Several studies have documented that administration of GH or somatostatin analogs to adults with GHD or GH excess, respectively, ameliorates abnormal biochemical profile and the associated somatic impairments. However, whether these improvements translate into improved physical function in adults with GHD or GH excess remains largely unknown, and their impact on HR-QoL controversial. Review of placebo-controlled trials to date suggests that GH and somatostatin analogs have greater effects on gas exchange and aerobic performance than as anabolic agents on skeletal muscle mass and function. Future investigations should include dose-response studies to establish the optimal combination of pharmacological agents plus exercise required to improve not only biochemical markers but also physical function and HR-QoL in adults with GHD or GH excess.
Collapse
Affiliation(s)
- Linda J Woodhouse
- School of Rehabilitation Science, McMaster University, Hamilton, Ontario, Canada
| | | | | | | |
Collapse
|
414
|
Eduardo Carreño J, Apablaza F, Paz Ocaranza M, E. Jalil J. Hipertrofia cardiaca: eventos moleculares y celulares. Rev Esp Cardiol 2006. [DOI: 10.1157/13087900] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
415
|
Carreño JE, Apablaza F, Ocaranza MP, Jalil JE. Cardiac Hypertrophy: Molecular and Cellular Events. ACTA ACUST UNITED AC 2006. [DOI: 10.1016/s1885-5857(06)60796-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
416
|
Venken K, De Gendt K, Boonen S, Ophoff J, Bouillon R, Swinnen JV, Verhoeven G, Vanderschueren D. Relative impact of androgen and estrogen receptor activation in the effects of androgens on trabecular and cortical bone in growing male mice: a study in the androgen receptor knockout mouse model. J Bone Miner Res 2006; 21:576-85. [PMID: 16598378 DOI: 10.1359/jbmr.060103] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
UNLABELLED The relative importance of AR and ER activation has been studied in pubertal male AR knockout and WT mice after orchidectomy and androgen replacement therapy, either with or without an aromatase inhibitor. AR activation dominates normal trabecular bone development and cortical bone modeling in male mice. Moreover, optimal periosteal bone expansion is only observed in the presence of both AR and ER activation. INTRODUCTION Androgen receptor (AR)-mediated androgen action has traditionally been considered a key determinant of male skeletal growth. Increasing evidence, however, suggests that estrogens are also essential for normal male bone growth. Therefore, the relative importance of AR-mediated and estrogen receptor (ER)-mediated androgen action after aromatization remains to be clarified. MATERIALS AND METHODS Trabecular and cortical bone was studied in intact or orchidectomized pubertal AR knockout (ARKO) and male wildtype (WT) mice, with or without replacement therapy (3-8 weeks of age). Nonaromatizable (dihydrotestosterone [DHT]) and aromatizable (testosterone [T]) androgens and T plus an aromatase inhibitor (anastrazole) were administered to orchidectomized ARKO and WT mice. Trabecular and cortical bone modeling were evaluated by static and dynamic histomorphometry, respectively. RESULTS AR inactivation or orchidectomy induced a similar degree of trabecular bone loss (-68% and -71%, respectively). Both DHT and T prevented orchidectomy-induced bone loss in WT mice but not in ARKO mice. Administration of an aromatase inhibitor did not affect T action on trabecular bone. AR inactivation and orchidectomy had similar negative effects on cortical thickness (-13% and -8%, respectively) and periosteal bone formation (-50% and -26%, respectively). In orchidectomized WT mice, both DHT and T were found to stimulate periosteal bone formation and, as a result, to increase cortical thickness. In contrast, the periosteum of ARKO mice remained unresponsive to either DHT or T. Interestingly, administration of an aromatase inhibitor partly reduced T action on periosteal bone formation in orchidectomized WT mice (-34% versus orchidectomized WT mice on T), but not in ARKO mice. This effect was associated with a significant decrease in serum IGF-I (-21% versus orchidectomized WT mice on T). CONCLUSIONS These findings suggest a major role for AR activation in normal development of trabecular bone and periosteal bone growth in male mice. Moreover, optimal stimulation of periosteal growth is only obtained in the presence of both AR and ER activation.
Collapse
Affiliation(s)
- Katrien Venken
- Laboratory for Experimental Medicine and Endocrinology, Katholieke Universiteit Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
417
|
Abstract
The liver is a primary target for the action of GH, a pituitary protein hormone that regulates a broad range of physiological processes, including long bone growth, fatty acid oxidation, glucose uptake, and hepatic steroid and foreign compound metabolism. GH exerts sex-dependent effects on the liver in many species, with many hepatic genes, most notably genes coding for cytochrome P450 (CYP) enzymes, being transcribed in a sex-dependent manner. Sex differences in CYP expression are most striking in rats and mice (up to 500-fold male-female differences), but are also seen, albeit to a much smaller degree, in humans, where they are an important determinant of the sex dependence of hepatic drug and steroid metabolism. This article examines the mechanisms whereby GH, via its sex-dependent temporal patterns of pituitary release, activates intracellular signaling leading to the sexually dimorphic transcription of CYPs and other liver-expressed genes. Recent findings implicating the GH-regulated transcription factor STAT5b (signal transducer and activator of transcription 5b), hepatocyte nuclear factors 3beta, 4alpha and 6, and sex differences in DNA methylation and chromatin structure in the sex-dependent actions of GH are reviewed, and current mechanistic models are evaluated.
Collapse
Affiliation(s)
- David J Waxman
- Department of Biology, Boston University, 5 Cummington Street, Boston, Massachusetts 02215, USA.
| | | |
Collapse
|
418
|
Masternak MM, Al-Regaiey KA, Del Rosario Lim MM, Jimenez-Ortega V, Panici JA, Bonkowski MS, Kopchick JJ, Wang Z, Bartke A. Caloric restriction and growth hormone receptor knockout: effects on expression of genes involved in insulin action in the heart. Exp Gerontol 2006; 41:417-29. [PMID: 16524678 PMCID: PMC3082456 DOI: 10.1016/j.exger.2006.01.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2005] [Revised: 01/20/2006] [Accepted: 01/24/2006] [Indexed: 01/16/2023]
Abstract
Blockade of growth hormone (GH), decreased insulin-like growth factor-1 (IGF1) action and increased insulin sensitivity are associated with life extension and an apparent slowing of the aging process. We examined expression of genes involved in insulin action, IR, IRS1, IRS2, IGF1, IGF1R, GLUT4, PPARs and RXRs in the hearts of normal and GHR-/- (KO) mice fed ad libitum or subjected to 30% caloric restriction (CR). CR increased the cardiac expression of IR, IRS1, IGF1, IGF1R and GLUT4 in normal mice and IRS1, GLUT4, PPARalpha and PPARbeta/delta in GHR-KO animals. Expression of IR, IRS1, IRS2, IGF1, GLUT4, PPARgamma and PPARalpha did not differ between GHR-KO and normal mice. These unexpected results suggest that CR may lead to major modifications of insulin action in the heart, but high insulin sensitivity of GHR-KO mice is not associated with alterations in the levels of most of the examined molecules related to intracellular insulin signaling.
Collapse
MESH Headings
- Aging/metabolism
- Animals
- Blotting, Western
- Caloric Restriction
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Gene Expression
- Glucose Transporter Type 4/genetics
- Glucose Transporter Type 4/metabolism
- Growth Hormone/genetics
- Growth Hormone/metabolism
- Insulin/metabolism
- Insulin Resistance
- Insulin-Like Growth Factor I/genetics
- Insulin-Like Growth Factor I/metabolism
- Male
- Mice
- Mice, Knockout
- Mice, Transgenic
- Myocardium/metabolism
- PPAR alpha/genetics
- PPAR alpha/metabolism
- PPAR delta/genetics
- PPAR delta/metabolism
- PPAR gamma/genetics
- PPAR gamma/metabolism
- PPAR-beta/genetics
- PPAR-beta/metabolism
- RNA, Messenger/analysis
- Receptor, IGF Type 1/genetics
- Receptor, IGF Type 1/metabolism
- Receptor, Insulin/genetics
- Receptor, Insulin/metabolism
- Receptors, Somatotropin/genetics
- Receptors, Somatotropin/metabolism
- Retinoid X Receptors/genetics
- Retinoid X Receptors/metabolism
- Reverse Transcriptase Polymerase Chain Reaction/methods
- Signal Transduction/physiology
Collapse
Affiliation(s)
- Michal M Masternak
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62794, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
419
|
Abstract
Cystic fibrosis (CF) is an autosomal recessive disease that results in lung failure and premature death. A long recognized symptom of CF is growth failure, which is clinically relevant because it correlates with the severity of lung disease. We describe growth retardation in a mouse model of CF and discuss its potential for modeling certain aspects of human growth retardation. Mice with a null mutation in Cftr (cystic fibrosis transmembrance conductance regulator) were compared with wild-type (WT) mice at 31, 45, and 84 d of age. CF mice were severely growth retarded in weight and length compared with wild-type controls. Serum insulin like growth factor I (Igf-1) was lower in CF mice by 31-55% (depending on age and sex) and it significantly correlated with the size of mice after controlling for gender, age, and Cftr genotype. There was a marginally significant deficiency of serum growth hormone (Gh) in CF females, but not males. Our findings were consistent with models of an energy deficit in rodents. We, therefore, assessed food intake and found no difference between CF and WT mice, suggesting that CF mice had a malabsorption-mediated energy deficit. We argue that CF mice are suited to study the effects of intestinal disease on growth as well as other proposed growth-modulating processes.
Collapse
Affiliation(s)
- Lewis A Rosenberg
- Department of Genetics, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | | | |
Collapse
|
420
|
Trejo JL, Carro E, Burks DJ. Experimental models for understanding the role of insulin-like growth factor-I and its receptor during development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2006; 567:27-53. [PMID: 16370135 DOI: 10.1007/0-387-26274-1_2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
|
421
|
Abstract
The growth hormone-insulin like growth factor (GH-IGF) axis plays a crucial role in the regulation of growth. Initially considered to be a mediator of growth hormone actions, IGF axis has been established as an independent endocrine system with wide array of actions. Recent advances have led to tremendous increase in the clinical utility of the IGF axis. IGF-based investigations (IGF1 and IGF binding protein 3) are now replacing GH-based investigations for evaluation and monitoring of disorders of the GH-IGF axis. IGF therapy has been successfully utilized in growth hormone insensitivity syndrome and GHD type 1B. The possibility of IGF axis as therapeutic options is being explored in wide variety of disorders like hypoxic-ischemic encephalopathy, Alzheimer's disease and psoriasis.
Collapse
Affiliation(s)
- Anurag Bajpai
- Department of Endocrinology and Diabetes, Center for Hormone Research, Royal Children's Hospital, Melbourne, Victoria, Australia
| | | |
Collapse
|
422
|
Smith FM, Garfield AS, Ward A. Regulation of growth and metabolism by imprinted genes. Cytogenet Genome Res 2006; 113:279-91. [PMID: 16575191 DOI: 10.1159/000090843] [Citation(s) in RCA: 136] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2005] [Accepted: 07/21/2005] [Indexed: 01/05/2023] Open
Abstract
A small sub-set of mammalian genes are subject to regulation by genomic imprinting such that only one parental allele is active in at least some sites of expression. Imprinted genes have diverse functions, notably including the regulation of growth. Much attention has been devoted to the insulin-like growth factor signalling pathway that has a major influence on fetal size and contains two components encoded by the oppositely imprinted genes, Igf2 (a growth promoting factor expressed from the paternal allele) and Igf2r (a growth inhibitory factor expressed from the maternal allele). These genes fit the parent-offspring conflict hypothesis for the evolution of genomic imprinting. Accumulated evidence indicates that at least one other fetal growth pathway exists that has also fallen under the influence of imprinting. It is clear that not all components of growth regulatory pathways are encoded by imprinted genes and instead it may be that within a pathway the influence of a single gene by each of the parental genomes may be sufficient for parent-offspring conflict to be enacted. A number of imprinted genes have been found to influence energy homeostasis and some, including Igf2 and Grb10, may coordinate growth with glucose-regulated metabolism. Since perturbation of fetal growth can be correlated with metabolic disorders in adulthood these imprinted genes are considered as candidates for involvement in this phenomenon of fetal programming.
Collapse
Affiliation(s)
- F M Smith
- Centre for Regenerative Medicine and Developmental Biology Programme, Department of Biology and Biochemistry, University of Bath, Bath, UK
| | | | | |
Collapse
|
423
|
Abstract
Insulin-like growth factors (IGFs) promote longitudinal growth and display anabolic effects in adult bone by acting through endocrine and autocrine/paracrine mechanisms. Binding of IGF-I to its specific tyrosine-kinase receptor leads to interaction with the intracellular proteins, insulin receptor substrate-1 and -2, and the activation of distinct intracellular signaling pathways. In cartilage, IGF-I regulates the differentiation of chondrocytes and stimulates the synthesis of components of the extracellular matrix. In bone tissue, IGF-I increases the function of the differentiated osteoblasts and mediates selected anabolic actions of parathyroid hormone. Genetically modified mice, in which selected components of the IGF system were targeted in a tissue-specific fashion, have documented that circulating IGF-I is essential for physiological skeletal growth and adult bone remodeling and that local autocrine/paracrine IGF-I activities are required for optimal trabecular bone mass and mineralization. Studies in humans have indicated a correlation between serum IGF-I levels and bone mineral density. However, there is little information on the use of IGF-I in patients with metabolic bone disease.
Collapse
Affiliation(s)
- Elisabetta Gazzerro
- a Dipartimento di Neuroscienze e Riabilitazione, Instituto Giannina, Gaslini, Genova, Italy.
| | | |
Collapse
|
424
|
Chia DJ, Ono M, Woelfle J, Schlesinger-Massart M, Jiang H, Rotwein P. Characterization of distinct Stat5b binding sites that mediate growth hormone-stimulated IGF-I gene transcription. J Biol Chem 2005; 281:3190-7. [PMID: 16339156 DOI: 10.1074/jbc.m510204200] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A key agent in the anabolic actions of growth hormone (GH) is insulin-like growth factor-I (IGF-I), a 70-amino acid secreted protein with direct effects on somatic growth and tissue maintenance and repair. GH rapidly and potently stimulates IGF-I gene transcription by mechanisms independent of new protein synthesis, and recent studies have linked the transcription factor Stat5b to a regulatory network connecting the activated GH receptor on the cell membrane to the IGF-I gene in the nucleus. Here we analyze two distinct conserved GH response elements in the rat IGF-I locus that contain paired Stat5b sites. Each response element binds Stat5b in vivo in a GH-dependent way, as assessed by chromatin immunoprecipitation assays, and consists of one high affinity and one lower affinity Stat5b site, as determined by both qualitative and quantitative protein-DNA binding studies. In biochemical reconstitution experiments, both response elements are able to mediate GH-stimulated and Stat5b-dependent transcription when fused to a reporter gene containing either the major IGF-I promoter or a minimal neutral promoter, although the paired Stat5b sites located in the second IGF-I intron were more than twice as effective as the response element that mapped approximately 73 kb 5' to the IGF-I exon 1. Taken together, our results define the initial molecular architecture of a complicated GH-regulated transcriptional pathway, and suggest that apparently redundant hormone response elements provide a mechanism for amplifying GH action at a physiologically important target gene.
Collapse
Affiliation(s)
- Dennis J Chia
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, Oregon 97239, USA
| | | | | | | | | | | |
Collapse
|
425
|
Venken K, Schuit F, Van Lommel L, Tsukamoto K, Kopchick JJ, Coschigano K, Ohlsson C, Movérare S, Boonen S, Bouillon R, Vanderschueren D. Growth without growth hormone receptor: estradiol is a major growth hormone-independent regulator of hepatic IGF-I synthesis. J Bone Miner Res 2005; 20:2138-49. [PMID: 16294267 DOI: 10.1359/jbmr.050811] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2005] [Revised: 07/29/2005] [Accepted: 08/04/2005] [Indexed: 01/07/2023]
Abstract
UNLABELLED The role of estrogens in the regulation of pubertal growth independently of GH and its receptor was studied in male mice with disrupted GHRKO. E(2) rescued skeletal growth rates in GHRKO associated with an increase in hepatic and serum IGF-I. These data show that E(2) rescues pubertal growth during GH resistance through a novel mechanism of GHR-independent stimulation of hepatic IGF-I production. INTRODUCTION Growth hormone (GH) and estrogen play a pivotal role in pubertal growth and bone mineral acquisition. Estrogens can affect GH secretion and thereby provide a GH-dependent mechanism for their effects on skeletal growth. It is presently unclear if or to what extent estrogens are able to regulate pubertal growth and bone mineral accrual independently of GH and its receptor. MATERIALS AND METHODS Estradiol (E(2); 0.03 mug/day by subcutaneous silastic implants) was administered to orchidectomized (ORX) male mice with disrupted GHR (GHRKO) and corresponding WTs during late puberty (6-10 weeks). Longitudinal and radial bone growth, IGF-I in serum and its expression in liver, muscle, and bone, and liver gene expression were studied by histomorphometry, RIA, RT-PCR, microarrays, and Western blotting, respectively. RESULTS E(2) stimulated not only longitudinal (femur length and growth plate thickness) and radial growth (cortical thickness and periosteal perimeter), but also rescued longitudinal and periosteal growth rates in ORX GHRKO, whereas no significant changes occurred in WT. E(2) thereby upregulated serum IGF-I and liver IGF-I synthesis (+21% and +52%, respectively) in ORX GHRKO, whereas IGF-I synthesis in femur or muscle was unaffected. Study of the underlying mechanism of the stimulation of hepatic IGF-I expression showed that E(2) restored downregulated receptor signaling systems, such as the estrogen receptor alpha and the prolactin receptor. E(2) thereby recovered the Janus kinase (JAK)/signal transducers and activators of transcription (STAT) pathway as evidenced by a significantly increased activation of the transcription factor STAT5 in ORX GHRKO. CONCLUSIONS Our data show a stimulation of skeletal growth through upregulation of hepatic IGF-I by a hormone other than GH. E(2) rescues pubertal skeletal growth during GH resistance through a novel mechanism of GHR-independent stimulation of IGF-I synthesis in the liver.
Collapse
Affiliation(s)
- Katrien Venken
- Laboratory for Experimental Medicine and Endocrinology, Katholieke Universiteit Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
426
|
O'Shea PJ, Bassett JHD, Sriskantharajah S, Ying H, Cheng SY, Williams GR. Contrasting Skeletal Phenotypes in Mice with an Identical Mutation Targeted to Thyroid Hormone Receptor α1 or β. Mol Endocrinol 2005; 19:3045-59. [PMID: 16051666 DOI: 10.1210/me.2005-0224] [Citation(s) in RCA: 89] [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
Thyroid hormone (T(3)) regulates bone turnover and mineralization in adults and is essential for skeletal development. Surprisingly, we identified a phenotype of skeletal thyrotoxicosis in T(3) receptor beta(PV) (TRbeta(PV)) mice in which a targeted frameshift mutation in TRbeta results in resistance to thyroid hormone. To characterize mechanisms underlying thyroid hormone action in bone, we analyzed skeletal development in TRalpha1(PV) mice in which the same PV mutation was targeted to TRalpha1. In contrast to TRbeta(PV) mice, TRalpha1(PV) mutants exhibited skeletal hypothyroidism with delayed endochondral and intramembranous ossification, severe postnatal growth retardation, diminished trabecular bone mineralization, reduced cortical bone deposition, and delayed closure of the skull sutures. Skeletal hypothyroidism in TRalpha1(PV) mutants was accompanied by impaired GH receptor and IGF-I receptor expression and signaling in the growth plate, whereas GH receptor and IGF-I receptor expression and signaling were increased in TRbeta(PV) mice. These data indicate that GH receptor and IGF-I receptor are physiological targets for T(3) action in bone in vivo. The divergent phenotypes observed in TRalpha1(PV) and TRbeta(PV) mice arise because the pituitary gland is a TRbeta-responsive tissue, whereas bone is TRalpha responsive. These studies provide a new understanding of the complex relationship between central and peripheral thyroid status.
Collapse
Affiliation(s)
- Patrick J O'Shea
- Molecular Endocrinology Group, 5th Floor Clinical Research Building, Medical Research Council Clinical Sciences Centre, Hammersmith Hospital, Du Cane Road, London W12 0NN, United Kingdom
| | | | | | | | | | | |
Collapse
|
427
|
Rowland JE, Kerr LM, White M, Noakes PG, Waters MJ. Heterozygote effects in mice with partial truncations in the growth hormone receptor cytoplasmic domain: assessment of growth parameters and phenotype. Endocrinology 2005; 146:5278-86. [PMID: 16166215 DOI: 10.1210/en.2005-0939] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The GH receptor (GHR) is essential for normal postnatal growth and development, and the molecular basis of GHR action has been studied intensively. Clinical case studies and more recently mouse models have revealed the extensive phenotype of impaired GH action. We recently reported two new mouse models, possessing cytoplasmic truncations at position 569 (plus Y539/545-F) and 391, which were created to identify functional subdomains within the cytoplasmic signaling domain. In the homozygous state, these animals show progressively impaired postnatal growth coupled with complex changes in gene expression. We describe here an extended phenotype analysis encompassing the heterozygote state to identify whether single copies of these mutant receptors bring about partial or dominant-negative phenotypes. It appears that the retention of the ubiquitin-dependent endocytosis motif in the N-terminal cytoplasmic domain permits turnover of these mutant receptors because no dominant-negative phenotype is seen. Nonetheless, we do observe partial impairment of postnatal growth in heterozygotes supporting limited haploinsufficiency. Reproductive function is impaired in these models in a progressive manner, in parallel with loss of signal transducer and activator of transcription-5 activation ability. In summary, we describe a more comprehensive phenotypic analysis of these mouse models, encompassing overall and longitudinal body growth, reproductive function, and hormonal status in both the heterozygote and homozygote state. Our results suggest that patients expressing single copies of similarly mutated GHRs would not display an obvious clinical phenotype.
Collapse
Affiliation(s)
- Jennifer E Rowland
- Institute for Molecular Bioscience, University of Queensland, St. Lucia, Australia
| | | | | | | | | |
Collapse
|
428
|
Alvaro D, Metalli VD, Alpini G, Onori P, Franchitto A, Barbaro B, Glaser SS, Francis H, Cantafora A, Blotta I, Attili AF, Gaudio E. The intrahepatic biliary epithelium is a target of the growth hormone/insulin-like growth factor 1 axis. J Hepatol 2005; 43:875-83. [PMID: 16083987 DOI: 10.1016/j.jhep.2005.04.011] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2004] [Revised: 03/11/2005] [Accepted: 04/07/2005] [Indexed: 02/08/2023]
Abstract
BACKGROUND/AIMS We evaluated the role and mechanisms by which the GH/IGF1 axis modulates cholangiocyte proliferation. METHODS GH-receptors (GH-R), IGF1, IGFBP3 (binding protein 3), IGF1-R and receptor substrates (IRS) were evaluated in cholangiocytes of normal or bile duct-ligated (BDL) rat livers. The effects of GH and IGF1 on proliferation of normal quiescent cholangiocytes and the transduction pathways involved were investigated. RESULTS IGF1, GH-R, IGF1-R, IRS-1/2 were expressed in normal cholangiocytes and overexpressed in cholangiocytes proliferating after BDL which also secrete IGF1 in a higher amount than normal cells. IGFBP3, which may counter-regulate IGF1 effects, was decreased in BDL cholangiocytes. IGF1 promoted cholangiocyte proliferation in association with overexpression of p-IGF1R, IRS1, IRS-2, p-ERK1/2 and p-AKT. GH induced IGF1 expression and release in isolated cholangiocytes, and reproduced the effects of IGF1 but GH effects were abolished by IGF1-R blocking antibody, suggesting IGF1 as a mediator of GH. Finally, IGF1 and 17beta-estradiol reciprocally potentiated their proliferative effects on cholangiocytes, and by interacting at both receptor and post-receptor levels. CONCLUSIONS Cholangiocytes respond to GH with production and release of IGF1 that modulates cell proliferation by transduction pathways involving IGF1-R, IRS1/2 and both ERK and PI3-kinase pathways. The biliary epithelium is a target of GH/IGF1 liver axis.
Collapse
Affiliation(s)
- Domenico Alvaro
- Division of Gastroenterology, University of Rome, La Sapienza, Rome, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
429
|
He K, Loesch K, Cowan JW, Li X, Deng L, Wang X, Jiang J, Frank SJ. Janus kinase 2 enhances the stability of the mature growth hormone receptor. Endocrinology 2005; 146:4755-4765. [PMID: 16081639 DOI: 10.1210/en.2005-0514] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The abundance of surface GH receptor (GHR) is an important determinant of cellular GH sensitivity and is regulated at both transcriptional and posttranscriptional levels. In previous studies of GHR-expressing Janus kinase 2 (JAK2)-deficient human fibrosarcoma cells (gamma2A-GHR), we demonstrated that stable transfection with JAK2 resulted in increased steady-state levels of mature GHR (endoH-resistant; relative molecular mass, 115-140 kDa) relative to precursor GHR (endoH-sensitive; relative molecular mass, 100 kDa). We now examine further the effects of JAK2 on GHR trafficking by comparing gamma2A-GHR to gamma2A-GHR cells stably reconstituted with JAK2 (C14 cells). In the presence of JAK2, GHR surface expression was increased, as assessed by surface biotinylation, 125I-labeled human GH cell surface binding, and immunofluorescence microscopy assays. Although the absence of JAK2 precluded GH-stimulated signaling, GH-induced GHR disulfide linkage (a proxy for the GH-induced conformational changes in the GHR dimer) proceeded independent of JAK2 expression, indicating that the earliest steps in GH-induced GHR triggering are not prevented by the absence of JAK2. RNA interference-mediated knockdown of JAK2 in C14 cells resulted in a decreased mature to precursor ratio, supporting a primary role for JAK2 either in enhancing GHR biogenesis or dampening mature GHR degradation. To address these potential mechanisms, metabolic pulse-chase labeling experiments and experiments in which the fate of previously synthesized GHR was followed by anti-GHR immunoblotting after cycloheximide treatment (cycloheximide chase experiments) were performed. These indicated that the presence of JAK2 conferred modest enhancement (1.3- to 1.5-fold) in GHR maturation but substantially prolonged the t1/2 of the mature GHR, suggesting a predominant effect on mature GHR stability. Cycloheximide chase experiments with metalloprotease, proteasome, and lysosome inhibitors indicated that the enhanced stability of mature GHR conferred by JAK2 is not related to effects on constitutive receptor metalloproteolysis but rather is a result of reduced constitutive endosomal/lysosomal degradation of the mature GHR. These results are discussed in the context of emerging information on how JAK-family members modulate surface expression of other cytokine receptors.
Collapse
Affiliation(s)
- Kai He
- Endocrinology Section Medical Service, Veterans Affairs Medical Center, and Department of Medicine, University of Alabama at Birmingham, 1530 3rd Avenue South, BDB 861, Birmingham, Alabama 35294-0012, USA
| | | | | | | | | | | | | | | |
Collapse
|
430
|
Stefan M, Ji H, Simmons RA, Cummings DE, Ahima RS, Friedman MI, Nicholls RD. Hormonal and metabolic defects in a prader-willi syndrome mouse model with neonatal failure to thrive. Endocrinology 2005; 146:4377-85. [PMID: 16002520 DOI: 10.1210/en.2005-0371] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Prader-Willi syndrome (PWS) has a biphasic clinical phenotype with failure to thrive in the neonatal period followed by hyperphagia and severe obesity commencing in childhood among other endocrinological and neurobehavioral abnormalities. The syndrome results from loss of function of several clustered, paternally expressed genes in chromosome 15q11-q13. PWS is assumed to result from a hypothalamic defect, but the pathophysiological basis of the disorder is unknown. We hypothesize that a fetal developmental abnormality in PWS leads to the neonatal phenotype, whereas the adult phenotype results from a failure in compensatory mechanisms. To address this hypothesis and better characterize the neonatal failure to thrive phenotype during postnatal life, we studied a transgenic deletion PWS (TgPWS) mouse model that shares similarities with the first stage of the human syndrome. TgPWS mice have fetal and neonatal growth retardation associated with profoundly reduced insulin and glucagon levels. Consistent with growth retardation, TgPWS mice have deregulated liver expression of IGF system components, as revealed by quantitative gene expression studies. Lethality in TgPWS mice appears to result from severe hypoglycemia after postnatal d 2 after depletion of liver glycogen stores. Consistent with hypoglycemia, TgPWS mice appear to have increased fat oxidation. Ghrelin levels increase in TgPWS reciprocally with the falling glucose levels, suggesting that the rise in ghrelin reported in PWS patients may be secondary to a perceived energy deficiency. Together, the data reveal defects in endocrine pancreatic function as well as glucose and hepatic energy metabolism that may underlie the neonatal phenotype of PWS.
Collapse
Affiliation(s)
- M Stefan
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia 19104-6140, USA
| | | | | | | | | | | | | |
Collapse
|
431
|
Niu T, Rosen CJ. The insulin-like growth factor-I gene and osteoporosis: a critical appraisal. Gene 2005; 361:38-56. [PMID: 16183214 DOI: 10.1016/j.gene.2005.07.016] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2005] [Revised: 06/16/2005] [Accepted: 07/08/2005] [Indexed: 12/17/2022]
Abstract
Osteoporosis, a disorder of skeletal fragility, is common in the elderly, and its prevalence is increasing as more individuals with low bone mineral density (BMD), the strongest predictor of fracture risk, are detected. Previous basic and clinical studies imply there is a significant role for insulin-like growth factor-I (IGF-I) in determining BMD. Recently, polymorphisms upstream of the P1 promoter region of the human IGF-I gene have been found to be associated with serum levels of IGF-I, BMD and fracture risk in various ethnic groups. Multiple quantitative trait loci (QTLs) have been identified that underlie serum IGF-I in a mouse intercross between two inbred strains. The most promising QTL on mouse chromosome 6 has provided clues for unraveling the molecular mechanisms that regulate osteoblast differentiation. Genomic engineering resulting in IGF-I deficient mice, and mice with targeted over-expression of IGF-I reinforce the essential role of IGF-I in bone development at both the embryonic and postnatal stages. Thus, it is apparent that significant new insights into the role of the IGF-I gene in bone remodeling occur through several distinct mechanisms: (1) the skeletal IGF regulatory system; (2) the systemic growth hormone/IGF-I axis; (3) parathyroid hormone signaling; (4) sex steroids; and (5) the OPG/RANKL/RANK cytokine system. Molecular dissection of the IGF regulatory system and its signaling pathway in bone may reveal novel therapeutic targets for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Tianhua Niu
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA.
| | | |
Collapse
|
432
|
Wang Y, Iordanov H, Swietlicki EA, Wang L, Fritsch C, Coleman T, Semenkovich CF, Levin MS, Rubin DC. Targeted intestinal overexpression of the immediate early gene tis7 in transgenic mice increases triglyceride absorption and adiposity. J Biol Chem 2005; 280:34764-75. [PMID: 16085642 DOI: 10.1074/jbc.m507058200] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Following loss of functional small bowel surface area due to surgical resection, the remnant gut undergoes an adaptive response characterized by increased crypt cell proliferation and enhanced villus height and crypt depth, resulting in augmented intestinal nutrient absorptive capacity. Previous studies showed that expression of the immediate early gene tis7 is markedly up-regulated in intestinal enterocytes during the adaptive response. To study its role in the enterocyte, transgenic mice were generated that specifically overexpress TIS7 in the gut. Nucleotides -596 to +21 of the rat liver fatty acid-binding protein promoter were used to direct abundant overexpression of TIS7 into small intestinal upper crypt and villus enterocytes. TIS7 transgenic mice had increased total body adiposity and decreased lean muscle mass compared with normal littermates. Oxygen consumption levels, body weight, surface area, and small bowel weight were decreased. On a high fat diet, transgenic mice exhibited a more rapid and proportionately greater gain in body weight with persistently elevated total body adiposity and increased hepatic fat accumulation. Bolus fat feeding resulted in a greater increase in serum triglyceride levels and an accelerated appearance of enterocytic, lamina propria, and hepatic fat. Changes in fat homeostasis were linked to increased expression of genes involved in enterocytic triglyceride metabolism and changes in growth with decreased insulin-like growth factor-1 expression. Thus, TIS7 overexpression in the intestine altered growth, metabolic rate, adiposity, and intestinal triglyceride absorption. These results suggest that TIS7 is a unique mediator of nutrient absorptive and metabolic adaptation following gut resection.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
433
|
Abstract
This review examines the rationale for targeting the insulin-like growth factor (IGF)-I receptor in the therapy of human tumours and their metastases. The rationale is based on two crucial findings: 1) in experimental animals, normal cells are only partially affected by the deletion of the IGF-I receptor, whereas tumour cells undergo apoptosis when the IGF-I receptor is downregulated; and 2) cells with a deleted IGF-I receptor are refractory to transformation by viral and cellular oncogenes. This review focuses on the mechanisms underlying the experimental findings, and discusses the possibility of extrapolating the results obtained in animals to the cure of human tumours.
Collapse
Affiliation(s)
- Renato Baserga
- Thomas Jefferson University, Kimmel Cancer Center, 233 S. 10th Street, 624 BLSB, Philadelphia, PA 19107, USA.
| |
Collapse
|
434
|
Hwa V, Little B, Adiyaman P, Kofoed EM, Pratt KL, Ocal G, Berberoglu M, Rosenfeld RG. Severe growth hormone insensitivity resulting from total absence of signal transducer and activator of transcription 5b. J Clin Endocrinol Metab 2005; 90:4260-6. [PMID: 15827093 DOI: 10.1210/jc.2005-0515] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT The central clinical feature of GH insensitivity (GHI) is severe growth failure associated with elevated serum concentrations of GH and abnormally low serum levels of IGF-I. GHI can be the result of an abnormality in the GH receptor or aberrancies downstream of the GH receptor. OBJECTIVE We investigated the GH-IGF-I axis in a young female GHI subject who presented with a height of 114 cm (-7.8 sd score) at age 16.4 yr. PATIENT The subject, from a consanguineous pedigree, had circulating levels of GH and GH-binding protein that were normal to elevated, whereas IGF-I (7.2 ng/ml; normal, 242-600), IGF-binding protein-3 (543 ng/ml; normal, 2500-4800), and acid-labile subunit (1.22 microg/ml; normal, 5.6-16) levels were abnormally low and failed to increase during an IGF-I generation test. DESIGN Dermal fibroblast cultures were established with the consent of the patient and family. Immunoblot analysis of cell lysates and DNA sequencing of her signal transducer and activator of transcription 5b (STAT5b), a critical intermediate of the GH-IGF-I axis, were performed. RESULTS Sequencing of the STAT5b gene revealed a novel homozygous insertion of a single nucleotide in exon 10. The insertion resulted in a frame shift, leading to early protein termination and consequent lack of immunodetectable STAT5b protein. CONCLUSION The identification of a second case of severe growth failure associated with STAT5b mutation implicates a unique and critical role for STAT5b in GH stimulation of IGF-I gene expression and statural growth.
Collapse
Affiliation(s)
- Vivian Hwa
- Department of Pediatrics, NRC5, Oregon Health & Sciences University, 3181 S.W. Sam Jackson Park Road, Portland, Oregon 97239-3098, USA.
| | | | | | | | | | | | | | | |
Collapse
|
435
|
Abstract
Forty years ago, when growth hormone (GH) first became available for use, the molecular basis for growth hormone deficiency (GHD) was entirely unknown. Despite continued difficulties with the biochemical diagnosis of GHD, we now accept the existence of underlying genetic abnormalities as the basis for disorders involving GH secretion. Similar challenges are encountered when one considers the causes of non-GHD short stature, namely idiopathic short stature (ISS). Categorization of the causes for ISS by insulin-like growth factor I (IGF-I) concentrations provides a basis for speculation about the potential for IGF-I gene polymorphisms or binding protein abnormalities influencing the development of ISS-related growth failure.
Collapse
Affiliation(s)
- Ron G Rosenfeld
- Lucile Packard Foundation for Children's Health, 770 Welch Road, Suite 350, Palo Alto, CA 94022, USA.
| |
Collapse
|
436
|
King D, Jarjoura D, McEwen HA, Askew MJ. Growth hormone injections improve bone quality in a mouse model of osteogenesis imperfecta. J Bone Miner Res 2005; 20:987-93. [PMID: 15883639 DOI: 10.1359/jbmr.050108] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2004] [Revised: 01/05/2005] [Accepted: 01/12/2005] [Indexed: 11/18/2022]
Abstract
UNLABELLED Systemic growth hormone injections increased spine and femur length in a mouse model of OI. Femur BMC, cross-sectional area, and BMD were increased. Smaller gains were produced in vertebral BMC and cross-sectional area. Biomechanical testing showed improvements to structural and material properties in the femur midshaft, supporting expanded testing of growth hormone therapy in children with OI. INTRODUCTION Osteoblasts in heterozygous Cola2oim mutant mice produce one-half the normal amounts of the alpha2 strand of type I procollagen. The mice experience a mild osteogenesis imperfecta (OI) phenotype, with femurs and vertebrae that require less force than normal to break in a biomechanical test. MATERIALS AND METHODS Subcutaneous injections of recombinant human growth hormone (rhGH) or saline were given 6 days per week to oim/+ mice between 3 and 12 weeks of age, in a protocol designed to simulate a trial on OI children. RESULTS rhGH injections promoted significant weight gain and skeletal growth compared with saline-treated control animals. Femur and spine lengths were increased significantly. Significant increases at the femur midshaft in cortical BMD (2.2%), BMC (15.5%), and cross-sectional area (13%) were produced by rhGH treatment. Increases in the same cortical bone parameters were measured in the metaphyseal region of the femur and in tail vertebrae, but lumbar vertebrae showed significant increases in BMC (9.6%) and cross-sectional area (10.1%) of trabecular bone. Three-point bending testing documented functional improvements to the femur mid-shafts. GH treatment produced significant increases in bone stiffness (23.7%), maximum load (30.8%), the energy absorbed by the femurs to the point of maximum load (44.5%), and the energy to actual fracture (40.4%). The ultimate stress endured by the bone material was increased by 14.1%. CONCLUSIONS Gains in bone length, cross-sectional area, BMD, BMC, structural biomechanical properties, and strength were achieved without directly addressing the genetic collagen defect in the mice. Results support expanded clinical testing of GH injections in children with OI.
Collapse
Affiliation(s)
- Donna King
- Department of Biochemistry and Molecular Pathology, Northeastern Ohio Universities College of Medicine, Rootstown, Ohio 44272, USA.
| | | | | | | |
Collapse
|
437
|
Shiura H, Miyoshi N, Konishi A, Wakisaka-Saito N, Suzuki R, Muguruma K, Kohda T, Wakana S, Yokoyama M, Ishino F, Kaneko-Ishino T. Meg1/Grb10 overexpression causes postnatal growth retardation and insulin resistance via negative modulation of the IGF1R and IR cascades. Biochem Biophys Res Commun 2005; 329:909-16. [PMID: 15752742 DOI: 10.1016/j.bbrc.2005.02.047] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2005] [Indexed: 11/25/2022]
Abstract
The Meg1/Grb10 protein has been implicated as an adapter protein in the signaling pathways from insulin receptor (IR) and insulin-like growth factor 1 receptor (IGF1R) in vitro. To elucidate its in vivo function, four independent Meg1/Grb10 transgenic mouse lines were established, and the effects of excess Meg1/Grb10 on both postnatal growth and glucose metabolism were examined. All of the Meg1/Grb10 transgenic mice showed growth retardation after weaning (3-4 weeks), which indicates that ectopic overexpression of Meg1/Grb10 inhibits postnatal growth that is mediated by IGF1 via IGF1R. In addition, the mice became hyperinsulinemic owing to high levels of insulin resistance, which demonstrates that Meg1/Grb10 also modulates the insulin receptor cascade negatively in vivo. Type II diabetes arose frequently in the two transgenic lines, which also showed impaired glucose tolerance. In these mice, severe atrophy of the pancreatic acinus cells was associated with high-level production of Meg1/Grb10 in the pancreas. These results suggest that Meg1/Grb10 inhibits the function of both insulin and IGF1 receptors in these cells, since a similar phenotype has been reported for Ir and Igf1r double knockout mice. Taken together, these results indicate that Meg1/Grb10 interacts with both insulin and IGF1 receptors in vivo, and negatively regulates the IGF growth pathways via these receptors.
Collapse
Affiliation(s)
- Hirosuke Shiura
- Department of Epigenetics, Medical Research Institute, Tokyo Medical and Dental University, 2-3-10 Kandasurugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
438
|
Wargelius A, Fjelldal PG, Benedet S, Hansen T, Björnsson BT, Nordgarden U. A peak in gh-receptor expression is associated with growth activation in Atlantic salmon vertebrae, while upregulation of igf-I receptor expression is related to increased bone density. Gen Comp Endocrinol 2005; 142:163-8. [PMID: 15862560 DOI: 10.1016/j.ygcen.2004.12.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2004] [Accepted: 12/14/2004] [Indexed: 11/26/2022]
Abstract
Growth hormone (GH) and insulin-like growth factor-I (IGF-I) play major roles in the endocrine regulation of fish growth, but their interdependency and mode of action has not been well elucidated. The GH-IGF-I system is essential for normal vertebral growth in mouse, but this has not been studied in fish. To study the interplay between GH, IGF-I, and their receptors, postsmolt Atlantic salmon were studied during spring growth (January-June 2003). From January to June, fish were sampled regularly for plasma and vertebral bone. The vertebra was collected from the same anterior-posterior position. The growth hormone receptor (ghr) (There is no determined nomenclature of salmon genes but we stick to the nomenclature which is consequent for zebrafish, where all gene names are named with small letters and in italic.) expression in the vertebrae peaked in the end of February coinciding with high levels of plasma GH and IGF-I, and an increase of vertebral growth rate. From April to June, plasma IGF-I levels decreased together with ghr expression in the vertebrae, while plasma GH did not decrease. In May and June, expression of the igf-I receptor (igf-Ir) increased 4- to 5-fold, which coincided with an increase in bone density. The changes seen in gene expression of the IGF-I and GH receptors suggest that these hormones are involved in vertebral growth and bone density.
Collapse
Affiliation(s)
- Anna Wargelius
- Institute of Marine Research, Matre, N-5984 Matredal, Norway.
| | | | | | | | | | | |
Collapse
|
439
|
Abstract
In broad terms, there are 3 types of cardiac hypertrophy: normal growth, growth induced by physical conditioning (i.e., physiologic hypertrophy), and growth induced by pathologic stimuli. Recent evidence suggests that normal and exercise-induced cardiac growth are regulated in large part by the growth hormone/IGF axis via signaling through the PI3K/Akt pathway. In contrast, pathological or reactive cardiac growth is triggered by autocrine and paracrine neurohormonal factors released during biomechanical stress that signal through the Gq/phospholipase C pathway, leading to an increase in cytosolic calcium and activation of PKC. Here we review recent developments in the area of these cardiotrophic kinases, highlighting the utility of animal models that are helping to identify molecular targets in the human condition.
Collapse
Affiliation(s)
- Gerald W Dorn
- Heart and Vascular Center, Medical Center, University of Cincinnati, Cincinnati, Ohio 45267-0542, USA.
| | | |
Collapse
|
440
|
Abstract
PURPOSE The pathophysiology of osteoporosis has seen many recent progress especially with the use of genetically modified animal models. CURRENT KNOWLEDGE AND KEY POINTS Among many discoveries, one can notice the crucial role of LRP5, GH, IGF-1 and the sex hormones receptors in the acquisition of the peak bone mass, the control of bone remodeling by the sympathetic nervous system and his implication as a transmitter of mechanical loading in bone. Also, the role of estrogen and androgen receptors as well as the aromatase is specified according to sexes. The role of growth plate's chondrocytes in the installation of the trabecular bone network is better and better demonstrated. The greater periosteal apposition in men, mediated by androgens receptor, seems to explain the greatest radial growth and so the greatest bone resistance to mechanical strains like a lower fracture rate in men compared to women. The bone microarchitecture and quality explain an important part of the mechanical properties of bones and why considering the same bone mass one bone is breaking and another one not. FUTURE PROSPECTS AND PROJECTS Many therapeutic applications should finalize the discovery of these new bone cells signalisation pathways.
Collapse
Affiliation(s)
- Régis Levasseur
- Service de rhumatologie, CHU de Caen, avenue de la Côte-de-Nacre, 14033 Caen, France
| | | | | |
Collapse
|
441
|
Easton RM, Cho H, Roovers K, Shineman DW, Mizrahi M, Forman MS, Lee VMY, Szabolcs M, de Jong R, Oltersdorf T, Ludwig T, Efstratiadis A, Birnbaum MJ. Role for Akt3/protein kinase Bgamma in attainment of normal brain size. Mol Cell Biol 2005; 25:1869-78. [PMID: 15713641 PMCID: PMC549378 DOI: 10.1128/mcb.25.5.1869-1878.2005] [Citation(s) in RCA: 471] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Studies of Drosophila and mammals have revealed the importance of insulin signaling through phosphatidylinositol 3-kinase and the serine/threonine kinase Akt/protein kinase B for the regulation of cell, organ, and organismal growth. In mammals, three highly conserved proteins, Akt1, Akt2, and Akt3, comprise the Akt family, of which the first two are required for normal growth and metabolism, respectively. Here we address the function of Akt3. Like Akt1, Akt3 is not required for the maintenance of normal carbohydrate metabolism but is essential for the attainment of normal organ size. However, in contrast to Akt1-/- mice, which display a proportional decrease in the sizes of all organs, Akt3-/- mice present a selective 20% decrease in brain size. Moreover, although Akt1- and Akt3-deficient brains are reduced in size to approximately the same degree, the absence of Akt1 leads to a reduction in cell number, whereas the lack of Akt3 results in smaller and fewer cells. Finally, mammalian target of rapamycin signaling is attenuated in the brains of Akt3-/- but not Akt1-/- mice, suggesting that differential regulation of this pathway contributes to an isoform-specific regulation of cell growth.
Collapse
Affiliation(s)
- Rachael M Easton
- Department of Medicine, University of Pennsylvania School of Medicine, Clinical Research Building 322, 415 Curie Blvd., Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
442
|
Abstract
The growth hormone/IGF-1-signaling (GH/IGF-1-signaling) system is involved in numerous physiological processes during normal growth and development and also in the aging process. Understanding the regulation of this system is therefore of importance to the biologist. Studies conducted over the past decade have shown that the JAK/STAT pathways are involved in GH signaling to the nucleus. More recently, evidence has been presented that a member of the SOCS family, SOCS2, is a negative regulator of GH signaling. This story began several years ago with the dramatic demonstration of gigantism in the SOCS2-knockout mouse. A more specific definition of the role of SOCS2 in GH signaling is provided in this issue of the JCI by the demonstration that the overgrowth phenotype of the SOCS2-/- mouse is dependent upon the presence of endogenous GH and that administration of GH to mice lacking both endogenous GH and SOCS2 produced excessive growth.
Collapse
Affiliation(s)
- Derek Leroith
- Diabetes Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Cancer Institute, NIH, Bethesda, Maryland 20892-1758, USA.
| | | |
Collapse
|
443
|
Stoffel W, Jenke B, Blöck B, Zumbansen M, Koebke J. Neutral sphingomyelinase 2 (smpd3) in the control of postnatal growth and development. Proc Natl Acad Sci U S A 2005; 102:4554-9. [PMID: 15764706 PMCID: PMC555473 DOI: 10.1073/pnas.0406380102] [Citation(s) in RCA: 131] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Neutral sphingomyelinases sphingomyelin phosphodiesterase (SMPD)2 and -3 hydrolyze sphingomyelin to phosphocholine and ceramide. smpd2 is expressed ubiquitously, and smpd3 is expressed predominantly in neurons of the CNS. Their activation and the functions of the released ceramides have been associated with signaling pathways in cell growth, differentiation, and apoptosis. However, these cellular responses remain poorly understood. Here we describe the generation and characterization of the smpd3(-/-) and smpd2(-/-)smpd3(-/-) double mutant mouse, which proved to be devoid of neutral sphingomyelinase activity. SMPD3 plays a pivotal role in the control of late embryonic and postnatal development: the smpd3-null mouse develops a novel form of dwarfism and delayed puberty as part of a hypothalamus-induced combined pituitary hormone deficiency. Our studies suggest that SMPD3 is segregated into detergent-resistant subdomains of Golgi membranes of hypothalamic neurosecretory neurons, where its transient activation modifies the lipid bilayer, an essential step in the Golgi secretory pathway. The smpd3(-/-) mouse might mimic a form of human combined pituitary hormone deficiency.
Collapse
Affiliation(s)
- Wilhelm Stoffel
- Laboratory of Molecular Neurosciences, Center of Molecular Medicine, Center of Biochemistry, Faculty of Medicine, University of Cologne, D-50931 Cologne, Germany.
| | | | | | | | | |
Collapse
|
444
|
Clark RG. Recombinant Human Insulin-Like Growth Factor I (IGF-I): Risks and Benefits of Normalizing Blood IGF-I Concentrations. Horm Res Paediatr 2005; 62 Suppl 1:93-100. [PMID: 15761240 DOI: 10.1159/000080766] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Recombinant human (rh) insulin-like growth factor I (IGF-I) is being developed as a therapy for short stature caused by IGF deficiency (IGFD) and also for diabetes mellitus. To complement the human efficacy and safety data, a large amount of information is available regarding the pharmacology and toxicology of rhIGF-I in animals. This review summarizes the risks and benefits of normalizing blood IGF-I concentrations in IGFD, especially with regard to carcinogenicity, and compares and contrasts safety data for rhIGF-I, recombinant human growth hormone (rhGH), and insulin. A major difference between rhIGF-I and rhGH is that rhIGF-I (like insulin) has hypoglycaemic activity, whereas rhGH opposes insulin action and is diabetogenic. In most of their actions, GH and IGF-I are similar. IGF-I mediates most of the actions of GH, so the safety of rhGH and that of rhIGF-I also share many common features. In animals, the transgenic expression of hGH has been shown to act directly, by activating the prolactin receptor, to increase the incidence of mammary and prostate tumours. In comparison, the over-expression of IGF-I in animals or the administration of rhIGF-I does not have a carcinogenic effect. In formal toxicology and carcinogenicity studies, rhIGF-I has similar effects to insulin in that it can increase food intake, body size, and the growth rate of existing tumours. In animals and humans, IGFD has many long-term detrimental effects besides short stature: it increases the risk of diabetes, cardiovascular disease, and low bone mineral density. Therefore, a case can be made for replacement therapy with rhIGF-I to normalize blood IGF-I levels and reverse the detrimental effects of IGFD.
Collapse
Affiliation(s)
- Ross G Clark
- Tercica, Inc., San Francisco, CA 94080-7111, USA.
| |
Collapse
|
445
|
Woelfle J, Chia DJ, Massart-Schlesinger MB, Moyano P, Rotwein P. Molecular physiology, pathology, and regulation of the growth hormone/insulin-like growth factor-I system. Pediatr Nephrol 2005; 20:295-302. [PMID: 15549418 DOI: 10.1007/s00467-004-1602-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2004] [Revised: 06/22/2004] [Accepted: 06/24/2004] [Indexed: 01/18/2023]
Abstract
Since the somatomedin hypothesis of growth hormone (GH) action was first formulated nearly 50 years ago, the key roles of both GH and insulin-like growth factor (IGF)-I in human growth have been confirmed and extended to include local effects on tissue maintenance and repair. More recent insights have revealed a dark side to the GH/IGF-I signaling system. Both proteins have been implicated as potential contributing factors in selected human cancers, and normal activity through this signaling pathway has been linked to diminished lifespan in experimental animals. This review highlights both the positive and negative aspects of the GH/IGF-I-growth pathway. The overall goal is to reinforce the need for more complete understanding of the mechanisms of signaling and action of GH and IGF-I, in order to separate, if possible, the potentially beneficial outcomes on growth and on tissue maintenance and repair from deleterious effects on cancer risk and lifespan.
Collapse
Affiliation(s)
- Joachim Woelfle
- Molecular Medicine Division, Department of Medicine, Oregon Health and Science University, Portland, Oregon 97239-3098, USA
| | | | | | | | | |
Collapse
|
446
|
Clark RG. Recombinant insulin-like growth factor-1 as a therapy for IGF-1 deficiency in renal failure. Pediatr Nephrol 2005; 20:290-4. [PMID: 15682316 DOI: 10.1007/s00467-004-1714-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2004] [Revised: 09/24/2004] [Accepted: 09/29/2004] [Indexed: 10/25/2022]
Abstract
Renal disease in children disrupts the growth hormone (GH) and insulin-like growth factor (IGF) axis and causes growth failure. Although GH therapy stimulates growth in these children, their short stature is likely due to a form of IGF-1 deficiency (IGFD) rather than GH deficiency. Recent experimental data have caused us to reconsider the importance of IGF-1 and IGFD to human growth. Pharmacology studies in rodents, as well as studies in patients with no functional GH receptors and primary IGFD, have shown that IGF-1 is an effective growth-promoting therapy. Gene knockout studies in mice have shown that IGF-1, rather than GH, is the major hormone controlling growth. In addition, both pharmacological and genetic studies have shown that there are effects of GH and IGF-1 that require their combined presence. In children with primary IGFD, where there is no GH signaling, recombinant human (rh)IGF-1 produces a large growth response, while in children who are GH and IGF-1 deficient, treatment with rhGH is the most-appropriate therapy. Children with short stature due to renal failure are GH sufficient and have some GH receptor signaling capacity, so that rhIGF-1, or rhIGF-1 plus rhGH, are logical therapeutic options and merit clinical testing.
Collapse
Affiliation(s)
- Ross G Clark
- Tercica, 651 Gateway Boulevard, Suite 950, South San Francisco, CA 94080, USA.
| |
Collapse
|
447
|
Tönshoff B, Kiepe D, Ciarmatori S. Growth hormone/insulin-like growth factor system in children with chronic renal failure. Pediatr Nephrol 2005; 20:279-89. [PMID: 15692833 DOI: 10.1007/s00467-005-1821-0] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2004] [Accepted: 12/30/2004] [Indexed: 10/25/2022]
Abstract
Disturbances of the somatotropic hormone axis play an important pathogenic role in growth retardation and catabolism in children with chronic renal failure (CRF). The apparent discrepancy between normal or elevated growth hormone (GH) levels and diminished longitudinal growth in CRF has led to the concept of GH insensitivity, which is caused by multiple alterations in the distal components of the somatotropic hormone axis. Serum levels of IGF-I and IGF-II are normal in preterminal CRF, while in end-stage renal disease (ESRD) IGF-I levels are slightly decreased and IGF-II levels slightly increased. In view of the prevailing elevated GH levels in ESRD, these serum IGF-I levels appear inadequately low. Indeed, there is both clinical and experimental evidence for decreased hepatic production of IGF-I in CRF. This hepatic insensitivity to the action of GH may be partly the consequence of reduced GH receptor expression in liver tissue and partly a consequence of disturbed GH receptor signaling. The actions and metabolism of IGFs are modulated by specific high-affinity IGFBPs. CRF serum has an IGF-binding capacity that is increased by seven- to tenfold, leading to decreased IGF bioactivity of CRF serum despite normal total IGF levels. Serum levels of intact IGFBP-1, -2, -4, -6 and low molecular weight fragments of IGFBP-3 are elevated in CRF serum in relation to the degree of renal dysfunction, whereas serum levels of intact IGFBP-3 are normal. Levels of immunoreactive IGFBP-5 are not altered in CRF serum, but the majority of IGFBP-5 is fragmented. Decreased renal filtration and increased hepatic production of IGFBP-1 and -2 both contribute to high levels of serum IGFBP. Experimental and clinical evidence suggests that these excessive high-affinity IGFBPs in CRF serum inhibit IGF action in growth plate chondrocytes by competition with the type 1 IGF receptor for IGF binding. These data indicate that growth failure in CRF is mainly due to functional IGF deficiency. Combined therapy with rhGH and rhIGF-I is therefore a logical approach.
Collapse
Affiliation(s)
- Burkhard Tönshoff
- University Children's Hospital, Im Neuenheimer Feld 153, 69120 Heidelberg, Germany.
| | | | | |
Collapse
|
448
|
van Buul-Offers SC, Smink JJ, Gresnigt R, Hamers N, Koedam J, Karperien M. Thyroid hormone, but not parathyroid hormone, partially restores glucocorticoid-induced growth retardation. Pediatr Nephrol 2005; 20:335-41. [PMID: 15688232 DOI: 10.1007/s00467-004-1690-y] [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] [Received: 06/02/2004] [Revised: 09/01/2004] [Accepted: 09/14/2004] [Indexed: 11/25/2022]
Abstract
Growth retardation is a serious side effect of long-term glucocorticoid (GC) treatment. In order to prevent or diminish this deleterious effect, a combination therapy including growth hormone (GH), a stimulator of bone growth, is often recommended. Parathyroid hormone (PTH) and thyroid hormone (T(4)) are important hormonal regulators of bone growth, and might also be helpful anabolic agents for counteracting the negative effects of GCs. Therefore, we studied the interaction of GCs in combination with a single dose of either PTH or T(4) on GC-induced growth retardation. Dexamethasone (Dex) treatment of mice for four weeks induced a significant growth inhibition of body length and weight and weights of several organs. PTH or T(4) alone did not affect the normal growth pattern. However, T(4) could partially restore the Dex-induced growth inhibition, whereas PTH could not. Although PTH did not affect total body growth, it did affect the height of the proliferative zone, which could be counteracted by Dex. This contrasts with T(4) treatment alone or in combination with Dex, which both resulted in a disturbed morphology of the growth plate. IGF-I mRNA, one of the mediators of longitudinal bone growth, was present in proliferative and hypertrophic chondrocytes. However, its expression was not affected by any of the treatments. In conclusion, T(4) but not PTH can partially counteract the effects of Dex on general body growth, with possible implications for future treatments of GC-induced growth retardation. Additionally, both T(4) and PTH, alone or in combination with Dex, have differential effects on the morphology of the growth plate.
Collapse
Affiliation(s)
- Sylvia C van Buul-Offers
- Department of Metabolic and Endocrine Diseases, University Medical Center Utrecht, P.O. Box 85090, 3508 AB, Utrecht, The Netherlands.
| | | | | | | | | | | |
Collapse
|
449
|
Sun LY, Evans MS, Hsieh J, Panici J, Bartke A. Increased neurogenesis in dentate gyrus of long-lived Ames dwarf mice. Endocrinology 2005; 146:1138-44. [PMID: 15564324 DOI: 10.1210/en.2004-1115] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Neurogenesis occurs throughout adult life in the dentate gyrus of mammalian hippocampus and has been suggested to play an important role in cognitive function. Multiple trophic factors including IGF-I have been demonstrated to regulate hippocampal neurogenesis. Ames dwarf mice live considerably longer than normal animals and maintain physiological function at youthful levels, including cognitive function, despite a deficiency of circulating GH and IGF-I. Here we show an increase in numbers of newly generated cells [bromodeoxyuridine (BrdU) positive] and newborn neurons (neuronal nuclear antigen and BrdU positive) in the dentate gyrus of adult dwarf mice compared with normal mice using BrdU labeling. Despite the profound suppression of hippocampal GH expression, hippocampal IGF-I protein levels are up-regulated and the corresponding mRNAs are as high in Ames dwarf as in normal mice. Our results suggest that local/hippocampal IGF-I expression may have induced the increase in hippocampal neurogenesis, and increased neurogenesis might contribute to the maintenance of youthful levels of cognitive function during aging in these long-lived animals.
Collapse
Affiliation(s)
- Liou Y Sun
- Geriatrics Research, Department of Internal Medicine, Southern Illinois University School of Medicine, Room 4389, 801 North Rutledge, Springfield, Illinois 62794-9628, USA
| | | | | | | | | |
Collapse
|
450
|
Pierce AL, Shimizu M, Beckman BR, Baker DM, Dickhoff WW. Time course of the GH/IGF axis response to fasting and increased ration in chinook salmon (Oncorhynchus tshawytscha). Gen Comp Endocrinol 2005; 140:192-202. [PMID: 15639147 DOI: 10.1016/j.ygcen.2004.10.017] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2004] [Revised: 10/14/2004] [Accepted: 10/22/2004] [Indexed: 10/26/2022]
Abstract
Body growth in vertebrates is chiefly regulated by the GH/IGF axis. Pituitary growth hormone (GH) stimulates liver insulin-like growth factor-I (IGF-I) production. During fasting, plasma IGF-I levels decline due to the development of liver GH resistance, while GH levels generally increase. In mammals, decreased insulin during fasting is thought to cause liver GH resistance. However, the sequence of events in the GH/IGF axis response to fasting is not well characterized, especially in non-mammalian vertebrates. We assessed the time course of the GH/IGF axis response to fasting and increased ration in chinook salmon. Fish were placed on Fasting, Increased, or Control rations, and sampled daily for 4 days and at more widely spaced intervals through 29 days. Plasma IGF-I, GH, insulin, and 41 kDa IGF binding protein (putative salmon IGFBP-3), and liver IGF-I gene expression were measured. Control and Increased ration fish did not differ strongly. Plasma IGF-I and 41 kDa IGFBP were significantly lower in Fasted versus Control fish from day 4 onward, and liver IGF-I gene expression was significantly lower from day 6 onward. Liver IGF-I gene expression and plasma IGF-I levels were correlated. Plasma insulin was lower in Fasted fish from day 6 onward. There was a trend toward increased GH in Fasted fish on days 1-2, and GH was significantly increased Fasted fish from day 3 onward. Fasted GH first increased (days 1-3) to a plateau of 10-20 ng/ml (days 4-12) and then increased dramatically (days 15-29), suggesting that the GH response to fasting had three phases. The early increase in GH, followed by the decrease in plasma IGF-I after 4 days, suggests that GH resistance developed within 4 days.
Collapse
Affiliation(s)
- A L Pierce
- School of Aquatic and Fishery Sciences, University of Washington, Seattle, WA 98195, USA.
| | | | | | | | | |
Collapse
|