301
|
Kaneko N, Nilsen TO, Tanaka H, Hara A, Shimizu M. Intact rather than total circulating insulin-like growth factor binding protein-1a is a negative indicator of growth in masu salmon. Am J Physiol Regul Integr Comp Physiol 2020; 318:R329-R337. [PMID: 31850820 DOI: 10.1152/ajpregu.00099.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Insulin-like growth factor binding protein (IGFBP)-1a is one of three major circulating forms in salmon and induced under catabolic conditions. However, there is currently no immunoassay available for this form because of a lack of standard and specific antibodies. We developed a time-resolved fluoroimmunoassay (TR-FIA) for salmon IGFBP-1a using recombinant protein for labeling, an assay standard, and production of antiserum. The TR-FIA had a low cross-reactivity (3.6%) with IGFBP-1b, another major form in the circulation. Fasting for 4 wk had no effect on serum immunoreactive (total) IGFBP-1a levels in yearling masu salmon, whereas 6-wk fasting significantly increased it. There was a significant, but weak, negative relationship between serum total IGFBP-1a level and individual growth rate (r2 = 0.12, P = 0.01). We next developed a ligand immuno-functional assay (LIFA) using europium-labeled IGF-I to quantify intact IGFBP-1a. In contrast to total IGFBP-1a, serum intact IGFBP-1a levels increased after 4 wk of fasting, and refeeding for 2 wk restored it to levels similar to those of the fed control. Serum intact IGFBP-1a levels showed a significant negative correlation with individual growth rate (r2 = 0.52, P < 0.001), which was as good as that of IGFBP-1b. Our findings using newly developed TR-FIA and LIFA suggest that regulation of intact IGFBP-1a levels has an important effect on growth in salmon and that intact IGFBP-1a is a negative index of salmon growth.
Collapse
Affiliation(s)
- Nobuto Kaneko
- Faculty of Fisheries Sciences, Hokkaido University, Hakodate, Hokkaido, Japan.,Norwegian Research Centre (NORCE) Environment, NORCE Norwegian Research Centre AS, Bergen, Norway
| | - Tom Ole Nilsen
- Norwegian Research Centre (NORCE) Environment, NORCE Norwegian Research Centre AS, Bergen, Norway.,Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Hanae Tanaka
- Faculty of Fisheries Sciences, Hokkaido University, Hakodate, Hokkaido, Japan
| | - Akihiko Hara
- Faculty of Fisheries Sciences, Hokkaido University, Hakodate, Hokkaido, Japan
| | - Munetaka Shimizu
- Faculty of Fisheries Sciences, Hokkaido University, Hakodate, Hokkaido, Japan
| |
Collapse
|
302
|
Strobel JS, Hack NL, Label KT, Cordova KL, Bersin TV, Journey ML, Beckman BR, Lema SC. Effects of food deprivation on plasma insulin-like growth factor-1 (Igf1) and Igf binding protein (Igfbp) gene transcription in juvenile cabezon (Scorpaenichthys marmoratus). Gen Comp Endocrinol 2020; 286:113319. [PMID: 31715138 DOI: 10.1016/j.ygcen.2019.113319] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/25/2019] [Accepted: 11/08/2019] [Indexed: 12/25/2022]
Abstract
The growth hormone (GH)/insulin-like growth factor (Igf) endocrine axis regulates somatic growth in the face of changing environmental conditions. In actinopterygian fishes, food availability is a key modulator of the somatotropic axis, with lower food intake generally depressing liver Igf1 release to diminish growth. Igf1 signaling, however, also involves several distinct IGF binding proteins (Igfbps), and the functional roles of many of these Igfbps in affecting growth during shifting food availability remain uncertain. Here, we tested how complete food deprivation (fasting) affected gene transcription for paralogs of all six types of Igfbps in the liver and fast-twitch skeletal muscle of cabezon (Scorpaenichthys marmoratus), a nearshore marine fish important for recreational fisheries in the eastern North Pacific Ocean. Juvenile cabezon were maintained as either fed (6% mass food⋅g fish wet mass-1⋅d-1) or fasted for 14 d. Fasted fish exhibited a lower body condition (K), a depressed mass-specific growth rate (SGR), and reduced plasma concentrations of Igf1. In the liver, fasting reduced the relative abundance of gene transcripts encoding Igfbps igfbp2a and igfbp2b, while significantly elevating mRNA levels for igfbp1a, igfbp1b, igfbp3b, and igfbp4. Fasting also reduced hepatic mRNA levels of GH receptor-1 (ghr1) - but not GH receptor-2 (ghr2) - supporting the idea that changes in liver sensitivity to GH may underlie the decline in plasma Igf1 during food deprivation. In skeletal muscle, fasting downregulated gene transcripts encoding igf1, igfbp2b, igfbp5b, and igfbp6b, while also upregulating mRNAs for igf2 and ghr2. These data demonstrate isoform-specific regulation of Igfbps in liver and skeletal muscle in cabezon experiencing food deprivation and reinforce the idea that the repertoire of duplicated Igfbp genes that evolved in actinopterygian fishes supports a diverse scope of endocrine and paracrine functions.
Collapse
Affiliation(s)
- Jackson S Strobel
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA
| | - Nicole L Hack
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA
| | - Kevin T Label
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA
| | - Kasey L Cordova
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA
| | - Theresa V Bersin
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA
| | - Meredith L Journey
- Lynker Technology, 202 Church St SE #536, Leesburg, VA 20175, Under Contract to Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Marine Fisheries Service, National Oceanic and Atmospheric Administration, Seattle Washington 98112, USA
| | - Brian R Beckman
- Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Marine Fisheries Service, National Oceanic and Atmospheric Administration, Seattle, Washington 98112, USA
| | - Sean C Lema
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA.
| |
Collapse
|
303
|
Nindl BC, Ahtiainen J, Gagnon SS, Taipale RS, Pierce JR, Martin BJ, Beckner ME, Lehti M, Häkkinen K, Kyröläinen H. Microdialysis-Assessed Exercised Muscle Reveals Localized and Differential IGFBP Responses to Unilateral Stretch Shortening Cycle Exercise. Front Endocrinol (Lausanne) 2020; 11:315. [PMID: 32547489 PMCID: PMC7272679 DOI: 10.3389/fendo.2020.00315] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/24/2020] [Indexed: 01/10/2023] Open
Abstract
Microdialysis allows for a preview into local muscle metabolism and can provide physiological insight that blood measurements cannot. Purpose: To examine the potential differential IGF-I system regulation in interstitial fluid during unilateral stretch shortening cycle exercise. Methods: 10 men (26 ± 7 year) performed unilateral jumping [stretch shortening cycle (SSC) exercise at 50% of optimal jump height] until volitional fatigue on a sled apparatus. Biological sampling took place using a catheter inserted into an antecubital vein (serum), and 100 kDa microdialysis probes inserted into the thigh muscle of each exercise/control leg (dialysate). Serum was drawn before (Pre; -3 h) and after SSC [Post I (+0 h), II (+3 h), or III (+20 h)]; dialysate was sampled for 2 h before (Pre), during/immediately after (Ex), and 3 h into recovery (Rec) following SSC. IGF-I system parameters (free/total IGF-I and IGFBPs 1-6) were measured with immunoassays. Interstitial free IGF-I was estimated from dialysate IGF-I and relative recovery (ethanol) correction. Data were analyzed with repeated measures ANOVA. Results: Serum total IGF-I remained elevated +3 h (Post II: 182.8 ± 37.6 vs. Pre: 168.3 ± 35.0 ng/mL, p < 0.01), but returned to baseline by +20 h (Post III vs. Pre, p = 0.31). No changes in serum free IGF-I were noted. Serum BP-1 and -3 increased over baseline, but not until + 20 h after SSC (Post III vs. Pre: 7.6 ± 4.9 vs. 3.7 ± 2.3 and 1,048.6 ± 269.2 vs. 891.4 ± 171.2 ng/mL, respectively). We observed a decreased serum BP-6 +3 h after SSC (p < 0.01), followed by a return to baseline at +20 h (p = 0.64 vs. Pre). There were no exercise-induced changes in serum BP-2, -4, or -5. Unlike serum, there were no changes in dialysate or interstitial free IGF-I in either leg (p > 0.05). Dialysate BP-1 remained increased in both exercise and control legs through 3 h into recovery (Rec vs. Pre, p < 0.01). Dialysate BP-3 also demonstrated a prolonged elevation over Pre SSC concentrations, but in the exercise leg only (Ex and Rec vs. Pre, p < 0.04). We observed a prolonged decrease in dialysate BP-5 (Ex and Rec vs. Pre, p < 0.03) and an increase in BP-4 IP in the exercise leg only. There were no changes relative to Pre SSC in dialysate BP-2 or -6. Conclusions: Unilateral exercise drives differential regulation of the IGF-I system at both local and systemic levels. More specifically, this is the first study to demonstrate that localized exercise increases IGFBP-3, IGFBP-4 and decreases in IGFBP-5 in muscle interstitial fluid.
Collapse
Affiliation(s)
- Bradley C. Nindl
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, University of Pittsburgh, Pittsburgh, PA, United States
- US Army Research Institute of Environmental Medicine, Natick, MA, United States
- Army Public Health Center, Aberdeen Proving Ground, MD, United States
- *Correspondence: Bradley C. Nindl
| | - Juha Ahtiainen
- Neuromuscular Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, Jyvaskyla, Finland
| | - Sheila S. Gagnon
- Neuromuscular Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, Jyvaskyla, Finland
| | - Ritva S. Taipale
- Neuromuscular Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, Jyvaskyla, Finland
| | - Joseph R. Pierce
- US Army Research Institute of Environmental Medicine, Natick, MA, United States
- Army Public Health Center, Aberdeen Proving Ground, MD, United States
| | - Brian J. Martin
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - Meaghan E. Beckner
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - M. Lehti
- Neuromuscular Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, Jyvaskyla, Finland
| | - Keijo Häkkinen
- Neuromuscular Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, Jyvaskyla, Finland
| | - Heikki Kyröläinen
- Neuromuscular Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, Jyvaskyla, Finland
| |
Collapse
|
304
|
Receptor Tyrosine Kinases in Osteosarcoma: 2019 Update. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1258:141-155. [PMID: 32767239 DOI: 10.1007/978-3-030-43085-6_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The primary conclusions of our 2014 contribution to this series were as follows: Multiple receptor tyrosine kinases (RTKs) likely contribute to aggressive phenotypes in osteosarcoma and, therefore, inhibition of multiple RTKs is likely necessary for successful clinical outcomes. Inhibition of multiple RTKs may also be useful to overcome resistance to inhibitors of individual RTKs as well as resistance to conventional chemotherapies. Different combinations of RTKs are likely important in individual patients. AXL, EPHB2, FGFR2, IGF1R, and RET were identified as promising therapeutic targets by our in vitro phosphoproteomic/siRNA screen of 42 RTKs in the highly metastatic LM7 and 143B human osteosarcoma cell lines. This chapter is intended to provide an update on these topics as well as the large number of osteosarcoma clinical studies of inhibitors of multiple tyrosine kinases (multi-TKIs) that were recently published.
Collapse
|
305
|
Duan C, Allard JB. Insulin-Like Growth Factor Binding Protein-5 in Physiology and Disease. Front Endocrinol (Lausanne) 2020; 11:100. [PMID: 32194505 PMCID: PMC7063065 DOI: 10.3389/fendo.2020.00100] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 02/17/2020] [Indexed: 12/25/2022] Open
Abstract
Insulin-like growth factor (IGF) signaling is regulated by a conserved family of IGF binding proteins (IGFBPs) in vertebrates. Among the six distinct types of IGFBPs, IGFBP-5 is the most highly conserved across species and has the broadest range of biological activities. IGFBP-5 is expressed in diverse cell types, and its expression level is regulated by a variety of signaling pathways in different contexts. IGFBP-5 can exert a range of biological actions including prolonging the half-life of IGFs in the circulation, inhibition of IGF signaling by competing with the IGF-1 receptor for ligand binding, concentrating IGFs in certain cells and tissues, and potentiation of IGF signaling by delivery of IGFs to the IGF-1 receptor. IGFBP-5 also has IGF-independent activities and is even detected in the nucleus. Its broad biological activities make IGFBP-5 an excellent representative for understanding IGFBP functions. Despite its evolutionary conservation and numerous biological activities, knockout of IGFBP-5 in mice produced only a negligible phenotype. Recent research has begun to explain this paradox by demonstrating cell type-specific and physiological/pathological context-dependent roles for IGFBP-5. In this review, we survey and discuss what is currently known about IGFBP-5 in normal physiology and human disease. Based on recent in vivo genetic evidence, we suggest that IGFBP-5 is a multifunctional protein with the ability to act as a molecular switch to conditionally regulate IGF signaling.
Collapse
|
306
|
Yang G, Zhao W, Qin C, Yang L, Meng X, Lu R, Yan X, Cao X, Zhang Y, Nie G. Igfbp3 in grass carp (Ctenopharyngodon idellus): Molecular identification and mRNA expression under glucose, insulin and glucagon. Comp Biochem Physiol B Biochem Mol Biol 2019; 242:110394. [PMID: 31866567 DOI: 10.1016/j.cbpb.2019.110394] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 12/11/2019] [Accepted: 12/17/2019] [Indexed: 12/16/2022]
Abstract
IGFBPs play a pivotal role in regulating the physiological function of IGFs (insulin-like growth factors). As an important member of IGFBPs, IGFBP3 is involved in the regulation of physiological functions of IGFs. To investigate the functional role of Igfbp3 in a herbivorous fish species, grass carp igfbp3 (GenBank accession no. MN251843) was isolated from the liver by molecular cloning. The ORF of grass carp igfbp3 was 882 bp, which encoded 293 amino acids, and the first 22 amino acids comprised the signal peptide. The predicted molecular weight of grass carp Igfbp3 is 31.95 kDa, and the theoretical isoelectric point is 8.32. Grass carp Igfbp3 displayed a high identity with its counterparts of common carp and zebrafish. And phylogenetic tree analysis showed that the grass carp Igfbp3 was clustered into the Igfbp3 subgroups of common carp and zebrafish. Tissue distribution study showed that igfbp3 was ubiquitously expressed in all tissues examined in the present study. High expression level of igfbp3 was detected in the heart, brain and fat of grass carp. The OGTT demonstrated that igfbp3 mRNA expression was significantly elevated in the liver of grass carp in response to glucose treatment. In vitro study showed that insulin could markedly stimulated igfbp3 mRNA expression in primary grass carp hepatocytes. Moreover, igfbp3 mRNA levels were also regulated by glucagon in grass carp primary hepatocytes. These results may provide the theoretical foundation for investigating the role of Igfbp3 in fish metabolism.
Collapse
Affiliation(s)
- Guokun Yang
- College of Fisheries, Henan Normal University, Xinxiang 453007, People's Republic of China; College of Fisheries, Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang 453007, People's Republic of China
| | - Wenli Zhao
- College of Fisheries, Henan Normal University, Xinxiang 453007, People's Republic of China; College of Fisheries, Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang 453007, People's Republic of China
| | - Chaobin Qin
- College of Fisheries, Henan Normal University, Xinxiang 453007, People's Republic of China; College of Fisheries, Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang 453007, People's Republic of China
| | - Liping Yang
- College of Fisheries, Henan Normal University, Xinxiang 453007, People's Republic of China; College of Fisheries, Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang 453007, People's Republic of China
| | - Xiaolin Meng
- College of Fisheries, Henan Normal University, Xinxiang 453007, People's Republic of China; College of Fisheries, Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang 453007, People's Republic of China
| | - Ronghua Lu
- College of Fisheries, Henan Normal University, Xinxiang 453007, People's Republic of China; College of Fisheries, Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang 453007, People's Republic of China
| | - Xiao Yan
- College of Fisheries, Henan Normal University, Xinxiang 453007, People's Republic of China; College of Fisheries, Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang 453007, People's Republic of China
| | - Xianglin Cao
- College of Fisheries, Henan Normal University, Xinxiang 453007, People's Republic of China; College of Fisheries, Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang 453007, People's Republic of China
| | - Yanmin Zhang
- College of Fisheries, Henan Normal University, Xinxiang 453007, People's Republic of China; College of Fisheries, Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang 453007, People's Republic of China
| | - Guoxing Nie
- College of Fisheries, Henan Normal University, Xinxiang 453007, People's Republic of China; College of Fisheries, Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang 453007, People's Republic of China.
| |
Collapse
|
307
|
Witman N, Zhou C, Grote Beverborg N, Sahara M, Chien KR. Cardiac progenitors and paracrine mediators in cardiogenesis and heart regeneration. Semin Cell Dev Biol 2019; 100:29-51. [PMID: 31862220 DOI: 10.1016/j.semcdb.2019.10.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/13/2019] [Accepted: 10/21/2019] [Indexed: 12/17/2022]
Abstract
The mammalian hearts have the least regenerative capabilities among tissues and organs. As such, heart regeneration has been and continues to be the ultimate goal in the treatment against acquired and congenital heart diseases. Uncovering such a long-awaited therapy is still extremely challenging in the current settings. On the other hand, this desperate need for effective heart regeneration has developed various forms of modern biotechnologies in recent years. These involve the transplantation of pluripotent stem cell-derived cardiac progenitors or cardiomyocytes generated in vitro and novel biochemical molecules along with tissue engineering platforms. Such newly generated technologies and approaches have been shown to effectively proliferate cardiomyocytes and promote heart repair in the diseased settings, albeit mainly preclinically. These novel tools and medicines give somehow credence to breaking down the barriers associated with re-building heart muscle. However, in order to maximize efficacy and achieve better clinical outcomes through these cell-based and/or cell-free therapies, it is crucial to understand more deeply the developmental cellular hierarchies/paths and molecular mechanisms in normal or pathological cardiogenesis. Indeed, the morphogenetic process of mammalian cardiac development is highly complex and spatiotemporally regulated by various types of cardiac progenitors and their paracrine mediators. Here we discuss the most recent knowledge and findings in cardiac progenitor cell biology and the major cardiogenic paracrine mediators in the settings of cardiogenesis, congenital heart disease, and heart regeneration.
Collapse
Affiliation(s)
- Nevin Witman
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Department of Medicine, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Chikai Zhou
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Niels Grote Beverborg
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Makoto Sahara
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Department of Medicine, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Department of Surgery, Yale University School of Medicine, CT, USA.
| | - Kenneth R Chien
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Department of Medicine, Karolinska Institutet, SE-171 77 Stockholm, Sweden.
| |
Collapse
|
308
|
Minakawa A, Fukuda A, Sato Y, Kikuchi M, Kitamura K, Wiggins RC, Fujimoto S. Podocyte hypertrophic stress and detachment precedes hyperglycemia or albuminuria in a rat model of obesity and type2 diabetes-associated nephropathy. Sci Rep 2019; 9:18485. [PMID: 31811176 PMCID: PMC6898392 DOI: 10.1038/s41598-019-54692-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 10/16/2019] [Indexed: 02/06/2023] Open
Abstract
Type2 diabetes-associated nephropathy is the commonest cause of renal failure. Mechanisms responsible are controversial. Leptin-deficient hyperphagic Zucker (fa/fa) rats were modeled to test the hypothesis that glomerular enlargement drives podocyte hypertrophic stress leading to accelerated podocyte detachment, podocyte depletion, albuminuria and progression. By 6weeks, prior to development of either hyperglycemia or albuminuria, fa/fa rats were hyperinsulinemic with high urinary IGF1/2 excretion, gaining weight rapidly, and had 1.6-fold greater glomerular volume than controls (P < 0.01). At this time the podocyte number per glomerulus was not yet reduced although podocytes were already hypertrophically stressed as shown by high podocyte phosphor-ribosomal S6 (a marker of mTORC1 activation), high urinary pellet podocin:nephrin mRNA ratio and accelerated podocyte detachment (high urinary pellet podocin:aquaporin2 mRNA ratio). Subsequently, fa/fa rats became both hyperglycemic and albuminuric. 24 hr urine albumin excretion correlated highly with decreasing podocyte density (R2 = 0.86), as a consequence of both increasing glomerular volume (R2 = 0.70) and decreasing podocyte number (R2 = 0.63). Glomerular podocyte loss rate was quantitatively related to podocyte detachment rate measured by urine pellet mRNAs. Glomerulosclerosis occurred when podocyte density reached <50/106um3. Reducing food intake by 40% to slow growth reduced podocyte hypertrophic stress and "froze" all elements of the progression process in place, but had small effect on hyperglycemia. Glomerular enlargement caused by high growth factor milieu starting in pre-diabetic kidneys appears to be a primary driver of albuminuria in fa/fa rats and thereby an under-recognized target for progression prevention. Progression risk could be identified prior to onset of hyperglycemia or albuminuria, and monitored non-invasively by urinary pellet podocyte mRNA markers.
Collapse
Affiliation(s)
- Akihiro Minakawa
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
- First Department of Internal Medicine, University of Miyazaki, Miyazaki, Japan
| | - Akihiro Fukuda
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan.
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Yufu, Japan.
| | - Yuji Sato
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Masao Kikuchi
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
- First Department of Internal Medicine, University of Miyazaki, Miyazaki, Japan
| | - Kazuo Kitamura
- First Department of Internal Medicine, University of Miyazaki, Miyazaki, Japan
| | - Roger C Wiggins
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Shouichi Fujimoto
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
- Department of Hemovascular Medicine and Artificial Organs, University of Miyazaki, Miyazaki, Japan
| |
Collapse
|
309
|
Celino-Brady FT, Petro-Sakuma CK, Breves JP, Lerner DT, Seale AP. Early-life exposure to 17β-estradiol and 4-nonylphenol impacts the growth hormone/insulin-like growth-factor system and estrogen receptors in Mozambique tilapia, Oreochromis mossambicus. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2019; 217:105336. [PMID: 31733503 PMCID: PMC6935514 DOI: 10.1016/j.aquatox.2019.105336] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 09/28/2019] [Accepted: 10/14/2019] [Indexed: 05/31/2023]
Abstract
It is widely recognized that endocrine disrupting chemicals (EDCs) released into the environment through anthropogenic activities can have short-term impacts on physiological and behavioral processes and/or sustained or delayed long-term developmental effects on aquatic organisms. While numerous studies have characterized the effects of EDCs on temperate fishes, less is known on the effects of EDCs on the growth and reproductive physiology of tropical species. To determine the long-term effects of early-life exposure to common estrogenic chemicals, we exposed Mozambique tilapia (Oreochromis mossambicus) yolk-sac fry to 17β-estradiol (E2) and nonylphenol (NP) and subsequently characterized the expression of genes involved in growth and reproduction in adults. Fry were exposed to waterborne E2 (0.1 and 1 μg/L) and NP (10 and 100 μg/L) for 21 days. After the exposure period, juveniles were reared for an additional 112 days until males were sampled. Gonadosomatic index was elevated in fish exposed to E2 (0.1 μg/L) while hepatosomatic index was decreased by exposure to NP (100 μg/L). Exposure to E2 (0.1 μg/L) induced hepatic growth hormone receptor (ghr) mRNA expression. The high concentration of E2 (1 μg/L), and both concentrations of NP, increased hepatic insulin-like growth-factor 1 (igf1) expression; E2 and NP did not affect hepatic igf2 and pituitary growth hormone (gh) levels. Both E2 (1 μg/L) and NP (10 μg/L) induced hepatic igf binding protein 1b (igfbp1b) levels while only NP (100 μg/L) induced hepatic igfbp2b levels. By contrast, hepatic igfbp6b was reduced in fish exposed to E2 (1 μg/L). There were no effects of E2 or NP on hepatic igfbp4 and igfbp5a expression. Although the expression of three vitellogenin transcripts was not affected, E2 and NP stimulated hepatic estrogen receptor (erα and erβ) mRNA expression. We conclude that tilapia exposed to E2 and NP as yolk-sac fry exhibit subsequent changes in the endocrine systems that control growth and reproduction during later life stages.
Collapse
Affiliation(s)
- Fritzie T Celino-Brady
- Department of Human Nutrition, Food and Animal Sciences, University of Hawai'i at Mānoa, 1955 East-West Road, Honolulu, HI 96822, USA.
| | - Cody K Petro-Sakuma
- Department of Human Nutrition, Food and Animal Sciences, University of Hawai'i at Mānoa, 1955 East-West Road, Honolulu, HI 96822, USA.
| | - Jason P Breves
- Department of Biology, Skidmore College, 815 N. Broadway, Saratoga Springs, NY 12866, USA.
| | - Darren T Lerner
- University of Hawai'i Sea Grant College Program, University of Hawai'i at Mānoa, 2525 Correa Road, Honolulu, HI 96822, USA.
| | - Andre P Seale
- Department of Human Nutrition, Food and Animal Sciences, University of Hawai'i at Mānoa, 1955 East-West Road, Honolulu, HI 96822, USA.
| |
Collapse
|
310
|
Safian D, Bogerd J, Schulz RW. Regulation of spermatogonial development by Fsh: The complementary roles of locally produced Igf and Wnt signaling molecules in adult zebrafish testis. Gen Comp Endocrinol 2019; 284:113244. [PMID: 31415728 DOI: 10.1016/j.ygcen.2019.113244] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/07/2019] [Accepted: 08/09/2019] [Indexed: 12/28/2022]
Abstract
Spermatogenesis is a cellular developmental process characterized by the coordinated proliferation and differentiation activities of somatic and germ cells in order to produce a large number of spermatozoa, the cellular basis of male fertility. Somatic cells in the testis, such as Leydig, peritubular myoid and Sertoli cells, provide structural and metabolic support and contribute to the regulatory microenvironment required for proper germ cell survival and development. The pituitary follicle-stimulating hormone (Fsh) is a major endocrine regulator of vertebrate spermatogenesis, targeting somatic cell functions in the testes. In fish, Fsh regulates Leydig and Sertoli cell functions, such as sex steroid and growth factor production, processes that also control the development of spermatogonia, the germ cell stages at the basis of the spermatogenic process. Here, we summarize recent advances in our understanding of mechanisms used by Fsh to regulate the development of spermatogonia. This involves discussing the roles of insulin-like growth factor (Igf) 3 and canonical and non-canonical Wnt signaling pathways. We will also discuss how these locally active regulatory systems interact to maintain testis tissue homeostasis.
Collapse
Affiliation(s)
- Diego Safian
- Reproductive Biology Group, Division Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, University of Utrecht, 3584 CH Utrecht, The Netherlands
| | - Jan Bogerd
- Reproductive Biology Group, Division Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, University of Utrecht, 3584 CH Utrecht, The Netherlands
| | - Rüdiger W Schulz
- Reproductive Biology Group, Division Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, University of Utrecht, 3584 CH Utrecht, The Netherlands; Reproduction and Developmental Biology Group, Institute of Marine Research, P.O. Box 1870, Nordnes, 5817 Bergen, Norway.
| |
Collapse
|
311
|
Khan S, Lu X, Huang Q, Tang J, Weng J, Yang Z, Lv M, Xu X, Xia F, Zhang M, Li Y, Liu S, Leng G, Spitzer N, Du J, Chen X. IGFBP2 Plays an Essential Role in Cognitive Development during Early Life. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1901152. [PMID: 31832311 PMCID: PMC6891907 DOI: 10.1002/advs.201901152] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 09/02/2019] [Indexed: 06/10/2023]
Abstract
Identifying the mechanisms underlying cognitive development in early life is a critical objective. The expression of insulin-like growth factor binding protein 2 (IGFBP2) in the hippocampus increases during neonatal development and is associated with learning and memory, but a causal connection has not been established. Here, it is reported that neurons and astrocytes expressing IGFBP2 are distributed throughout the hippocampus. IGFBP2 enhances excitatory inputs onto CA1 pyramidal neurons, facilitating intrinsic excitability and spike transmission, and regulates plasticity at excitatory synapses in a cell-type specific manner. It facilitates long-term potentiation (LTP) by enhancing N-methyl-d-aspartate (NMDA) receptor-dependent excitatory postsynaptic current (EPSC), and enhances neurite proliferation and elongation. Knockout of igfbp2 reduces the numbers of pyramidal cells and interneurons, impairs LTP and cognitive performance, and reduces tonic excitation of pyramidal neurons that are all rescued by IGFBP2. The results provide insight into the requirement for IGFBP2 in cognition in early life.
Collapse
Affiliation(s)
- Shumsuzzaman Khan
- Institute of NeuroscienceDepartment of Neurobiology, and Department of Neurology of Second Affiliated HospitalNHC and CAMS Key Laboratory of Medical NeurobiologyKey Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
- Present address:
Case Western Reserve UniversityClevelandOhioUSA
| | - Xinjiang Lu
- Institute of NeuroscienceDepartment of Neurobiology, and Department of Neurology of Second Affiliated HospitalNHC and CAMS Key Laboratory of Medical NeurobiologyKey Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
| | - Qingyao Huang
- Institute of NeuroscienceDepartment of Neurobiology, and Department of Neurology of Second Affiliated HospitalNHC and CAMS Key Laboratory of Medical NeurobiologyKey Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
| | - Jiawei Tang
- Institute of NeuroscienceDepartment of Neurobiology, and Department of Neurology of Second Affiliated HospitalNHC and CAMS Key Laboratory of Medical NeurobiologyKey Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
| | - Jian Weng
- Institute of NeuroscienceDepartment of Neurobiology, and Department of Neurology of Second Affiliated HospitalNHC and CAMS Key Laboratory of Medical NeurobiologyKey Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
| | - Zhi Yang
- Institute of NeuroscienceDepartment of Neurobiology, and Department of Neurology of Second Affiliated HospitalNHC and CAMS Key Laboratory of Medical NeurobiologyKey Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
| | - Minchao Lv
- Institute of NeuroscienceDepartment of Neurobiology, and Department of Neurology of Second Affiliated HospitalNHC and CAMS Key Laboratory of Medical NeurobiologyKey Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
| | - Xiaokang Xu
- Institute of NeuroscienceDepartment of Neurobiology, and Department of Neurology of Second Affiliated HospitalNHC and CAMS Key Laboratory of Medical NeurobiologyKey Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
| | - Fangyuan Xia
- Institute of NeuroscienceDepartment of Neurobiology, and Department of Neurology of Second Affiliated HospitalNHC and CAMS Key Laboratory of Medical NeurobiologyKey Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
| | - Mengchen Zhang
- Institute of NeuroscienceDepartment of Neurobiology, and Department of Neurology of Second Affiliated HospitalNHC and CAMS Key Laboratory of Medical NeurobiologyKey Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
| | - Yi Li
- Institute of NeuroscienceDepartment of Neurobiology, and Department of Neurology of Second Affiliated HospitalNHC and CAMS Key Laboratory of Medical NeurobiologyKey Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
| | - Shuangshuang Liu
- Institute of NeuroscienceDepartment of Neurobiology, and Department of Neurology of Second Affiliated HospitalNHC and CAMS Key Laboratory of Medical NeurobiologyKey Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
| | - Gareth Leng
- Experimental PhysiologyUniversity of EdinburghEdinburghEH8 9XDUK
| | | | - Jizeng Du
- Institute of NeuroscienceDepartment of Neurobiology, and Department of Neurology of Second Affiliated HospitalNHC and CAMS Key Laboratory of Medical NeurobiologyKey Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
| | - Xuequn Chen
- Institute of NeuroscienceDepartment of Neurobiology, and Department of Neurology of Second Affiliated HospitalNHC and CAMS Key Laboratory of Medical NeurobiologyKey Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
| |
Collapse
|
312
|
Kaneko N, Journey ML, Neville CM, Trudel M, Beckman BR, Shimizu M. Utilization of an endocrine growth index, insulin-like growth factor binding protein (IGFBP)-1b, for postsmolt coho salmon in the Strait of Georgia, British Columbia, Canada. FISH PHYSIOLOGY AND BIOCHEMISTRY 2019; 45:1867-1878. [PMID: 31297680 DOI: 10.1007/s10695-019-00681-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/02/2019] [Indexed: 06/10/2023]
Abstract
Monitoring the growth of salmon during their early marine phase provides insights into prey availability, and growth rates may be linked to risks of size-dependent mortality. However, the measurement of growth rate is challenging for free-living salmon in the ocean. Insulin-like growth factor (IGF)-I is a growth-promoting hormone that is emerging as a useful index of growth in salmon. In addition, laboratory-based studies using coho salmon have shown that one of circulating IGF-binding proteins (IGFBPs), IGFBP-1b, is induced by fasting and thus could be used as an inverse index of growth and/or catabolic state in salmon. However, few studies have measured plasma levels of IGFBP-1b in salmon in the wild. We measured plasma IGFBP-1b levels for postsmolt coho salmon collected in the Strait of Georgia and surrounding waters, British Columbia, Canada, and compared regional differences in IGFBP-1b to ecological information such as seawater temperature and stomach fullness. Plasma IGFBP-1b levels were the highest in fish from Eastern Johnstone Strait and relatively high in Queen Charlotte Strait and Western Johnstone Strait, which was in good agreement with the poor ocean conditions for salmon hypothesized to occur in that region. The molar ratio of plasma IGF-I to IGFBP-1b, a theoretical parameter of IGF-I availability to the receptor, discriminated differences among regions better than IGF-I or IGFBP-1b alone. Our data suggest that plasma IGFBP-1b reflects catabolic status in postsmolt coho salmon, as highlighted in fish in Eastern Johnston Strait, and is a useful tool to monitor negative aspects of salmon growth in the ocean.
Collapse
Affiliation(s)
- Nobuto Kaneko
- Faculty of Fisheries Sciences, Hokkaido University, 3-1-1 Minato, Hakodate, Hokkaido, 041-8611, Japan
| | - Meredith L Journey
- Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Marine Fisheries Service, 2725 Montlake Boulevard East, Seattle, WA, 98112, USA
| | - Chrys M Neville
- Fisheries and Oceans Canada, St Andrews Biological Station, St Andrews, New Brunswick, Canada
| | - Marc Trudel
- Fisheries and Oceans Canada, St Andrews Biological Station, St Andrews, New Brunswick, Canada
| | - Brian R Beckman
- Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Marine Fisheries Service, 2725 Montlake Boulevard East, Seattle, WA, 98112, USA
| | - Munetaka Shimizu
- Faculty of Fisheries Sciences, Hokkaido University, 3-1-1 Minato, Hakodate, Hokkaido, 041-8611, Japan.
| |
Collapse
|
313
|
Crim1 C140S mutant mice reveal the importance of cysteine 140 in the internal region 1 of CRIM1 for its physiological functions. Mamm Genome 2019; 30:329-338. [PMID: 31776724 DOI: 10.1007/s00335-019-09822-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 11/20/2019] [Indexed: 10/25/2022]
Abstract
Cysteine-rich transmembrane bone morphogenetic protein regulator 1 (CRIM1) is a type I transmembrane protein involved in the organogenesis of many tissues via its interactions with growth factors including BMP, TGF-β, and VEGF. In this study, we used whole-exome sequencing and linkage analysis to identify a novel Crim1 mutant allele generated by ENU mutagenesis in mice. This allele is a missense mutation that causes a cysteine-to-serine substitution at position 140, and is referred to as Crim1C140S. In addition to the previously reported phenotypes in Crim1 mutants, Crim1C140S homozygous mice exhibited several novel phenotypes, including dwarfism, enlarged seminal vesicles, and rectal prolapse. In vitro analyses showed that Crim1C140S mutation affected the formation of CRIM1 complexes and decreased the amount of the overexpressed CRIM1 proteins in the cell culture supernatants. Cys140 is located in the internal region 1 (IR1) of the N-terminal extracellular region of CRIM1 and resides outside any identified functional domains. Inference of the domain architecture suggested that the Crim1C140S mutation disturbs an intramolecular disulfide bond in IR1, leading to the protein instability and the functional defects of CRIM1. Crim1C140S highlights the functional importance of the IR1, and Crim1C140S mice should serve as a valuable model for investigating the functions of CRIM1 that are unidentified as yet.
Collapse
|
314
|
Werner H, Sarfstein R, Bruchim I. Investigational IGF1R inhibitors in early stage clinical trials for cancer therapy. Expert Opin Investig Drugs 2019; 28:1101-1112. [PMID: 31731883 DOI: 10.1080/13543784.2019.1694660] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: The insulin-like growth factors (IGFs) are a family of secreted peptide hormones with important roles in different cellular and organism functions. The biological activities of the IGFs are mediated by the IGF1 receptor (IGF1R), a cell surface, tyrosine kinase-containing heterotetramer that is linked to numerous cytoplasmic signaling cascades. The IGF1R displays potent antiapoptotic, pro-survival capacities and plays a key role in malignant transformation. Research has identified the IGF1R as a candidate therapeutic target in cancer.Areas covered: We offer a synopsis of ongoing efforts to target the IGF axis for therapeutic purposes. Our review includes a digest of early experimental work that led to the identification of IGF1R as a candidate therapeutic target in oncology.Expert opinion: Targeting of the IGF axis has yielded disappointing results in phase III trials, but it is important to learn from this to improve future trials in a rational manner. The potential of anti-IGF1R antibodies and small molecular weight inhibitors, alone or in combination with chemotherapy or other biological agents, should be investigated further in randomized studies. Moreover, the implementation of predictive biomarkers for patient selection will improve the outcome of future trials. Emerging personalized medicine could have a major impact on IGF1R targeting.
Collapse
Affiliation(s)
- Haim Werner
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Yoran Institute for Human Genome Research, Tel Aviv University, Tel Aviv, Israel
| | - Rive Sarfstein
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ilan Bruchim
- Gynecologic Oncology Division, Hillel Yaffe Medical Center, Technion Institute of Technology, Haifa, Israel
| |
Collapse
|
315
|
Khan S. IGFBP-2 Signaling in the Brain: From Brain Development to Higher Order Brain Functions. Front Endocrinol (Lausanne) 2019; 10:822. [PMID: 31824433 PMCID: PMC6883226 DOI: 10.3389/fendo.2019.00822] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 11/08/2019] [Indexed: 12/13/2022] Open
Abstract
Insulin-like growth factor-binding protein-2 (IGFBP-2) is a pleiotropic polypeptide that functions as autocrine and/or paracrine growth factors. IGFBP-2 is the most abundant of the IGFBPs in the cerebrospinal fluid (CSF), and developing brain showed the highest expression of IGFBP-2. IGFBP-2 expressed in the hippocampus, cortex, olfactory lobes, cerebellum, and amygdala. IGFBP-2 mRNA expression is seen in meninges, blood vessels, and in small cell-body neurons (interneurons) and astrocytes. The expression pattern of IGFBP-2 is often developmentally regulated and cell-specific. Biological activities of IGFBP-2 which are independent of their abilities to bind to insulin-like growth factors (IGFs) are mediated by the heparin binding domain (HBD). To execute IGF-independent functions, some IGFBPs have shown to bind with their putative receptors or to translocate inside the cells. Thus, IGFBP-2 functions can be mediated both via insulin-like growth factor receptor-1 (IGF-IR) and independent of IGF-Rs. In this review, I suggest that IGFBP-2 is not only involved in the growth, development of the brain but also with the regulation of neuronal plasticity to modulate high-level cognitive operations such as spatial learning and memory and information processing. Hence, IGFBP-2 serves as a neurotrophic factor which acts via metaplastic signaling from embryonic to adult stages.
Collapse
|
316
|
Zhang X, Zhang Z, Yu Z, Li J, Chen S, Sun R, Jia C, Zhu F, Meng Q, Xu S. Molecular cloning and expression pattern of IGFBP-2a in black porgy (Acanthopagrus schlegelii) and evolutionary analysis of IGFBP-2s in the species of Perciformes. FISH PHYSIOLOGY AND BIOCHEMISTRY 2019; 45:1731-1745. [PMID: 31418102 DOI: 10.1007/s10695-019-00665-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 05/24/2019] [Indexed: 06/10/2023]
Abstract
Insulin-like growth factor-binding protein-2 (IGFBP-2) plays a key role in regulating growth and development by its affinity with insulin-like growth factors (IGFs). In this study, we cloned the coding sequence (CDS) of IGFBP-2a from the black porgy (Acanthopagrus schlegelii) muscle and identified that the full-length CDS of IGFBP-2a was 882 bp. Real-time quantitative PCR revealed that IGFBP-2a was most abundant in the liver of the black porgy and backcross breed (F1♀×black porgy♂) but remained lower in each tested tissue in self-cross breed (F1♀×F1♂). In addition, the IGFBP-2a expression in the liver of three breeds showed a negative correlation with their growth rates, indicating that the IGFBP-2a played a growth-inhibiting role in the three breeds. We further identified 810 bp IGFBP-2b gene from the draft genome of black porgy. Finally, we examined the IGFBP-2a and IGFBP-2b genes by scanning the genomes of the species of Perciformes and found the IGFBP-2 gene duplication took place earlier than the divergence of perciform species. Interestingly, six positively selected sites were detected in both Perciformes IGFBP-2 genes, although both genes were identified to be under purifying selection. Specially, these positively selected sites were located in the functional domains, suggesting these sites played key roles in the growth of Perciformes. Our study partially explains the molecular basis for the prepotency in black porgy hybrids, which will provide guidance for their cultivation in the future.
Collapse
Affiliation(s)
- Xinyi Zhang
- Marine Fisheries Research Institute of Jiangsu Province, Nantong, 226007, China
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
| | - Zhiyong Zhang
- Marine Fisheries Research Institute of Jiangsu Province, Nantong, 226007, China
| | - Zhenpeng Yu
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
| | - Jiayi Li
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
| | - Shuyin Chen
- Marine Fisheries Research Institute of Jiangsu Province, Nantong, 226007, China.
| | - Ruijian Sun
- Marine Fisheries Research Institute of Jiangsu Province, Nantong, 226007, China
| | - Chaofeng Jia
- Marine Fisheries Research Institute of Jiangsu Province, Nantong, 226007, China
| | - Fei Zhu
- Marine Fisheries Research Institute of Jiangsu Province, Nantong, 226007, China
| | - Qian Meng
- Marine Fisheries Research Institute of Jiangsu Province, Nantong, 226007, China
| | - Shixia Xu
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, China.
| |
Collapse
|
317
|
Nowak M, Rehrauer H, Ay SS, Findik M, Boos A, Kautz E, Kowalewski MP. Gene expression profiling of the canine placenta during normal and antigestagen-induced luteolysis. Gen Comp Endocrinol 2019; 282:113194. [PMID: 31145892 DOI: 10.1016/j.ygcen.2019.05.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/23/2019] [Accepted: 05/26/2019] [Indexed: 12/20/2022]
Abstract
The domestic dog is the only domestic animal species that does not produce steroids in the placenta and instead relies on luteal steroids throughout pregnancy. Nevertheless, the canine placenta is highly responsive to steroids, and withdrawal of progesterone (P4) affects the feto-maternal unit, initializing the parturition cascade. Similar effects can be observed during antigestagen-induced abortion. Here, aiming to provide new insights into mechanisms involved in the termination of canine pregnancy, next generation sequencing (NGS, RNA-seq) was applied. Placental transcriptomes derived from natural prepartum and antigestagen-induced abortions were analyzed and compared with fully developed mid-gestation placentas. The contrast "prepartum luteolysis over mid-gestation" revealed 1973 differentially expressed genes (DEG). Terms associated with apoptosis, impairment of vascular function and activation of signaling of several cytokines (e.g., IL-8, IL-3, TGF-β) were overrepresented at natural luteolysis. When compared with mid-term, antigestagen treatment revealed 135 highly regulated DEG that were involved in the induced luteolysis and showed similar associations with functional terms and expression patterns as during natural luteolysis. The contrast "antigestagen-induced luteolysis over prepartum luteolysis" revealed that, although similar changes occur in both conditions, they are more pronounced during natural prepartum. Among P4-regulated DEG were those related to immune system and cortisol metabolism. It appears that, besides inducing placental PGF2α output, both natural and induced P4 withdrawal is associated with disruption of the feto-maternal interface, leading to impaired vascular functions, apoptosis and controlled modulation of the immune response. The time-related maturation of the feto-maternal interface needs to be considered because it may be clinically relevant.
Collapse
Affiliation(s)
- Marta Nowak
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich (UZH), Zurich, Switzerland
| | - Hubert Rehrauer
- Functional Genomics Center Zurich (FGCZ), ETH Zurich/University of Zurich, Zurich, Switzerland
| | - Serhan S Ay
- Department of Obstetrics and Gynaecology, Faculty of Veterinary Medicine, University of Ondokuz Mayis, Samsun, Turkey
| | - Murat Findik
- Department of Obstetrics and Gynaecology, Faculty of Veterinary Medicine, University of Ondokuz Mayis, Samsun, Turkey
| | - Alois Boos
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich (UZH), Zurich, Switzerland
| | - Ewa Kautz
- Department of Large Animal Diseases with Clinic, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (SGGW), Warsaw, Poland
| | - Mariusz P Kowalewski
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich (UZH), Zurich, Switzerland.
| |
Collapse
|
318
|
Joksić G, Tričković JF, Joksić I. Potential of Gentiana lutea for the Treatment of Obesity-associated Diseases. Curr Pharm Des 2019; 25:2071-2076. [PMID: 31538881 DOI: 10.2174/1381612825666190708215743] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 06/24/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND Obesity, diabetes, and associated diseases are increasing all over the world, and pose a great burden on public health. According to the latest reports, 440 million people are suffering from diabetes. Diabetes is caused by impaired ability to produce or respond to the hormone insulin consequently resulting in hyperglycemia. METHODS Data used for this review was obtained by using PUBMED/MEDLINE (1987-2018). The main data search terms were: Gentiana lutea, Gentiana lutea extract, Gentiana lutea constituents, obesity, diabetes mellitus, diabetic complications. RESULTS In the present review, we describe the potential of root powder of yellow gentian (Gentiana lutea) for the prevention of obesity and diabetes including complications related to this disease. CONCLUSION Reasonably effective, low-cost alternatives could fulfill an important role for a large part of the human population and could be of great value for the food market. Even a modest reduction of morbidity and mortality with respect to this disease translates into millions of lives saved.
Collapse
Affiliation(s)
- Gordana Joksić
- Vinca Institute of Nuclear Science, University of Belgrade, M.Petrovica Alasa 12-14, 11000 Belgrade, Serbia
| | - Jelena Filipović Tričković
- Vinca Institute of Nuclear Science, University of Belgrade, M.Petrovica Alasa 12-14, 11000 Belgrade, Serbia
| | - Ivana Joksić
- Vinca Institute of Nuclear Science, University of Belgrade, M.Petrovica Alasa 12-14, 11000 Belgrade, Serbia.,Clinic for Gynecology and Obstetrics Narodni Front, Kraljice Natalije 62, 11000, Belgrade, Serbia
| |
Collapse
|
319
|
Li J, Wang Y, Kang T, Li X, Niu C. Insulin-like growth factor binding proteins inhibit oocyte maturation of zebrafish. Gen Comp Endocrinol 2019; 281:83-90. [PMID: 31170402 DOI: 10.1016/j.ygcen.2019.06.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/27/2019] [Accepted: 06/02/2019] [Indexed: 12/16/2022]
Abstract
The function of insulin-like growth factor (Igf) system in ovary has attracted much attention, but the role of Igf binding proteins (Igfbps) in ovary is still largely unknown. In this study, the role of Igfbps in oocyte maturation was investigated in zebrafish. The expression of all eight identified Igfbps except Igfbp6b could be detected in the adult ovary and exhibited differential expression profiles during folliculogenesis. The expression of several Igfbps is dynamically changed during oocyte maturation induced by human chorionic gonadotropin (hCG). By treatment of an Igfbps inhibitor NBI-31772 in vitro, the oocyte maturation could be stimulated in a clear dose-, time- and stage-dependent manner. Such effects were also observed by administration of NBI-31772 in vivo. Igfbps are differentially expressed in both follicular cells and oocytes, but the effect of NBI-31772 could only be found in intact follicles and not in the denuded oocytes. Previous studies have demonstrated that Igf3 is the major Igf member in regulating oocyte maturation of zebrafish. Interestingly, NBI-31772 could increase the effect of Igf3 on oocyte maturation. Furthermore, we found the effect of NBI-31772 on oocyte maturation could be blocked by an Igf type 1 receptor inhibitor BMS-536924 in vitro, suggesting the Igfbps can inhibit the oocyte maturation via Igf/Igf1r pathway. Together, we provided the first evidence in fish that Igfbps inhibit oocyte maturation of zebrafish.
Collapse
Affiliation(s)
- Jianzhen Li
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu 730070, China.
| | - Yamei Wang
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu 730070, China
| | - Tao Kang
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu 730070, China
| | - Xuehui Li
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu 730070, China
| | - Caiyan Niu
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu 730070, China
| |
Collapse
|
320
|
Yang G, Chen B, Sun C, Yuan X, Zhang Y, Qin J, Li W. Molecular identification of grouper Igfbp1 and its mRNA expression in primary hepatocytes under Gh and insulin. Gen Comp Endocrinol 2019; 281:137-144. [PMID: 31176753 DOI: 10.1016/j.ygcen.2019.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 05/29/2019] [Accepted: 06/02/2019] [Indexed: 12/17/2022]
Abstract
The insulin-like growth factor (IGF) system plays a pivotal role in the regulation of growth, and IGF binding proteins (IGFBPs) are important regulatory factors in the IGF system. Generally, IGFBPs inhibit IGF actions by preventing its binding to receptors. Under some conditions, the IGFBPs can also enhance IGF actions. IGFBP1 is generally inhibitory to IGFI. In this study, the grouper (Epinephelus coioides) igfbp1 (MK621003) gene was cloned from the liver. The sequence of igfbp1 cDNA was 1055 bp and contained a 5'UTR of 127 bp and a 3'UTR of 247 bp, and the ORF of grouper igfbp1 was 741 bp, encoding 246 amino acids. The tissue distribution results showed that igfbp1 has a higher expression in the liver. In the nutritional status experiment, igfbp1 expression was significantly increased in the liver after 7 days of fasting and was markedly decreased after refeeding. In in vitro experiments, igfbp1 expression in grouper primary hepatocytes was significantly inhibited by recombinant grouper Gh (growth hormone) in a dose-dependent manner. Additionally, igfbp1 expression decreased in grouper primary hepatocytes upon incubation with insulin. This is the first report describing grouper igfbp1, and these findings contribute to understanding the roles of IGFBP1 in metabolism and growth in grouper.
Collapse
Affiliation(s)
- Guokun Yang
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Beichen Chen
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Caiyun Sun
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Xi Yuan
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yazhou Zhang
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Jingkai Qin
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Wensheng Li
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China.
| |
Collapse
|
321
|
Bøtkjær JA, Noer PR, Oxvig C, Yding Andersen C. A common variant of the pregnancy-associated plasma protein-A (PAPPA) gene encodes a protein with reduced proteolytic activity towards IGF-binding proteins. Sci Rep 2019; 9:13231. [PMID: 31519945 PMCID: PMC6744435 DOI: 10.1038/s41598-019-49626-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 07/05/2019] [Indexed: 02/03/2023] Open
Abstract
Pregnancy-associated plasma protein-A (PAPP-A) is a key regulator of insulin-like growth factor (IGF) bioactivity, by releasing the IGFs from their corresponding IGF-binding proteins (IGFBPs). The minor allele of the single nucleotide polymorphism (SNP), rs7020782 (serine < tyrosine), in PAPPA has previously been associated with recurrent pregnancy loss as well as with significant reduced levels of PAPP-A protein in human ovarian follicles. The aim of the present study was to reveal a possible functional effect of the rs7020782 SNP in PAPPA by comparing recombinant PAPP-A proteins from transfected human embryonic kidney 293 T cells. The proteolytic cleavage of IGFBP-4 was shown to be affected by the rs7020782 SNP in PAPPA, showing a significantly reduced cleavage rate for the serine variant compared to the tyrosine variant (p-value < 0.001). The serine variant also showed a trend towards reduced cleavage rates, that was not significant, towards IGFBP-2 and IGFBP-5 compared to the tyrosine variant. No differences were found when analysing cell surface binding, complex formation between PAPP-A and STC2 or proMBP, nor when analysing STC1 inhibition of PAPP-A-mediated IGFBP-4 cleavage. Regulation of IGF bioactivity in reproductive tissues is important and the rs7020782 SNP in PAPPA may disturb this regulation by altering the specific activity of PAPP-A.
Collapse
Affiliation(s)
- Jane Alrø Bøtkjær
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Rigshospitalet, Copenhagen University Hospital, Copenhagen University, Copenhagen, DK-2100, Denmark.
| | - Pernille Rimmer Noer
- Department of Molecular Biology and Genetics, University of Aarhus, Aarhus, DK-8000, Denmark
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, University of Aarhus, Aarhus, DK-8000, Denmark
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Rigshospitalet, Copenhagen University Hospital, Copenhagen University, Copenhagen, DK-2100, Denmark
| |
Collapse
|
322
|
Hack NL, Cordova KL, Glaser FL, Journey ML, Resner EJ, Hardy KM, Beckman BR, Lema SC. Interactions of long-term food ration variation and short-term fasting on insulin-like growth factor-1 (IGF-1) pathways in copper rockfish (Sebastes caurinus). Gen Comp Endocrinol 2019; 280:168-184. [PMID: 31022390 DOI: 10.1016/j.ygcen.2019.04.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/13/2019] [Accepted: 04/21/2019] [Indexed: 12/26/2022]
Abstract
Variation in food intake affects somatic growth by altering the expression of hormones in the somatotropic endocrine axis including insulin-like growth factor-1 (IGF-1). Here, we examined IGF-1 pathway responses to long- and short-term variation in food availability in copper rockfish (Sebastes caurinus), a nearshore Pacific rockfish important for commercial and recreational fisheries. Juvenile copper rockfish were raised under differing ration amounts (3% or 9% mass feed·g-1 fish wet mass·day-1) for 140 d to simulate 'long-term' feeding variation, after which some fish from both rations were fasted for 12 d to generate 'short-term' conditions of food deprivation. Rockfish on the 9% ration treatment grew more quickly than those on the 3% ration and were larger in mass, length, and body condition (k) after 152 d. Fish on the 9% ration had higher blood glucose than those on the 3% ration, with fasting decreasing blood glucose in both ration treatments, indicating that both long-term and short-term feed treatments altered energy status. Plasma IGF-1 was higher in rockfish from the 9% ration than those in the 3% ration and was also higher in fed fish than fasted fish. Additionally, plasma IGF-1 related positively to individual variation in specific growth rate (SGR). The positive association between IGF-1 and SGR showed discordance in fish that had experienced different levels of food and growth over the long-term but not short-term, suggesting that long-term nutritional experience can influence the relationship between IGF-1 and growth in this species. Rockfish on the 3% ration showed a lower relative abundance of gene transcripts encoding igf1 in the liver, but higher hepatic mRNAs for IGF binding proteins igfbp1a and igfbp1b. Fasting similarly decreased the abundance of igf1 mRNAs in the liver of fish reared under both the 9% and 3% rations, while concurrently increasing mRNAs encoding the IGF binding proteins igfbp1a, -1b, and -3a. Hepatic mRNAs for igfbp2b, -5a, and -5b were lower with long-term ration variation (3% ration) and fasting. Fish that experienced long-term reduced rations also had higher mRNA levels for igfbp3a, -3b, and IGF receptors isoforms A (igf1rA) and B (igf1rB) in skeletal muscle, but lower mRNA levels for igf1. Fasting increased muscle mRNA abundance for igfbp3a, igf1rA, and igf1rB, and decreased levels for igfbp2a and igf1. These data show that a positive relationship between circulating IGF-1 and individual growth rate is maintained in copper rockfish even when that growth variation relates to differences in food consumption across varying time scales, but that long- and short-term variation in food quantity can shift basal concentrations of circulating IGF-1 in this species.
Collapse
Affiliation(s)
- Nicole L Hack
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA
| | - Kasey L Cordova
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA
| | - Frances L Glaser
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA
| | - Meredith L Journey
- Lynker Technology, 202 Church St SE #536, Leesburg, VA 20175, Under Contract to Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Marine Fisheries Service, National Oceanic and Atmospheric Administration, Seattle, WA 98112, USA
| | - Emily J Resner
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA
| | - Kristin M Hardy
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA
| | - Brian R Beckman
- Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Marine Fisheries Service, National Oceanic and Atmospheric Administration, Seattle, WA, 98112, USA
| | - Sean C Lema
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA.
| |
Collapse
|
323
|
Pilitsi E, Peradze N, Perakakis N, Mantzoros CS. Circulating levels of the components of the GH/IGF-1/IGFBPs axis total and intact IGF-binding proteins (IGFBP) 3 and IGFBP 4 and total IGFBP 5, as well as PAPPA, PAPPA2 and Stanniocalcin-2 levels are not altered in response to energy deprivation and/or metreleptin administration in humans. Metabolism 2019; 97:32-39. [PMID: 31103608 DOI: 10.1016/j.metabol.2019.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 05/12/2019] [Indexed: 10/26/2022]
Abstract
OBJECTIVE It remains unclear whether food deprivation induces changes in components of the GH/IGF-1/IGFBPs axis and if yes, which ones are mediated by leptin, an adipocyte secreted hormone regulating neuroendocrine response to energy deprivation in animals and humans. We aimed to investigate components of the axis that have not been studied to date, i.e. IGF-binding proteins (IGFBPs) and related proteases (total and intact IGFBP 3 and IGFBP 4, total IGFBP 5, PAPPA, PAPPA2 and Stanniocalcin-2), during acute (short-term fasting in healthy subjects) and chronic (women with hypothalamic amenorrhea [HA] due to excessive exercise) energy deprivation and whether metreleptin administration, in replacement, supraphysiologic or pharmacologic levels, may mediate any changes of circulating levels of the above molecules in healthy individuals and in women with hypothalamic amenorrhea. METHODS We studied: 1) 11 healthy men and women during three four day admissions i.e. a baseline admission in the fed isocaloric state and two admissions in the complete food deprivation state for 72-h with either placebo (resulting in a hypoleptinemic state) or metreleptin administration in doses designed to normalize circulating leptin levels for the duration of the study, 2) 15 healthy men and women during three 72-hour long admissions in a complete food deprivation state receiving three escalating doses of metreleptin designed to bring circulating leptin levels to physiologic, supraphysiologic, or pharmacologic levels, and 3) 18 women with HA randomized to either metreleptin treatment in replacement doses or placebo for nine months. RESULTS There were no significant changes in the circulating profiles of the above molecules in the fasting vs. fed state and/or with metreleptin administration during acute and chronic energy deprivation. CONCLUSIONS The studied components of the GH/IGF-1/IGFBPs axis are not affected by energy deprivation, leptin deficiency associated with energy deprivation, or by metreleptin administration in physiologic, supraphysiologic or pharmacologic doses.
Collapse
Affiliation(s)
- Eleni Pilitsi
- Division of Endocrinology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA 02215, USA
| | - Natia Peradze
- Division of Endocrinology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA 02215, USA.
| | - Nikolaos Perakakis
- Division of Endocrinology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA 02215, USA
| | - Christos S Mantzoros
- Division of Endocrinology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA 02215, USA; Section of Endocrinology, VA Boston Healthcare System, Jamaica Plain, MA, USA
| |
Collapse
|
324
|
The wonders of BMP9: From mesenchymal stem cell differentiation, angiogenesis, neurogenesis, tumorigenesis, and metabolism to regenerative medicine. Genes Dis 2019; 6:201-223. [PMID: 32042861 PMCID: PMC6997590 DOI: 10.1016/j.gendis.2019.07.003] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/07/2019] [Accepted: 07/10/2019] [Indexed: 12/15/2022] Open
Abstract
Although bone morphogenetic proteins (BMPs) initially showed effective induction of ectopic bone growth in muscle, it has since been determined that these proteins, as members of the TGF-β superfamily, play a diverse and critical array of biological roles. These roles include regulating skeletal and bone formation, angiogenesis, and development and homeostasis of multiple organ systems. Disruptions of the members of the TGF-β/BMP superfamily result in severe skeletal and extra-skeletal irregularities, suggesting high therapeutic potential from understanding this family of BMP proteins. Although it was once one of the least characterized BMPs, BMP9 has revealed itself to have the highest osteogenic potential across numerous experiments both in vitro and in vivo, with recent studies suggesting that the exceptional potency of BMP9 may result from unique signaling pathways that differentiate it from other BMPs. The effectiveness of BMP9 in inducing bone formation was recently revealed in promising experiments that demonstrated efficacy in the repair of critical sized cranial defects as well as compatibility with bone-inducing bio-implants, revealing the great translational promise of BMP9. Furthermore, emerging evidence indicates that, besides its osteogenic activity, BMP9 exerts a broad range of biological functions, including stem cell differentiation, angiogenesis, neurogenesis, tumorigenesis, and metabolism. This review aims to summarize our current understanding of BMP9 across biology and the body.
Collapse
|
325
|
Gehrig JL, Venkatesh S, Chang HW, Hibberd MC, Kung VL, Cheng J, Chen RY, Subramanian S, Cowardin CA, Meier MF, O'Donnell D, Talcott M, Spears LD, Semenkovich CF, Henrissat B, Giannone RJ, Hettich RL, Ilkayeva O, Muehlbauer M, Newgard CB, Sawyer C, Head RD, Rodionov DA, Arzamasov AA, Leyn SA, Osterman AL, Hossain MI, Islam M, Choudhury N, Sarker SA, Huq S, Mahmud I, Mostafa I, Mahfuz M, Barratt MJ, Ahmed T, Gordon JI. Effects of microbiota-directed foods in gnotobiotic animals and undernourished children. Science 2019; 365:eaau4732. [PMID: 31296738 PMCID: PMC6683325 DOI: 10.1126/science.aau4732] [Citation(s) in RCA: 282] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 04/24/2019] [Accepted: 06/07/2019] [Indexed: 12/16/2022]
Abstract
To examine the contributions of impaired gut microbial community development to childhood undernutrition, we combined metabolomic and proteomic analyses of plasma samples with metagenomic analyses of fecal samples to characterize the biological state of Bangladeshi children with severe acute malnutrition (SAM) as they transitioned, after standard treatment, to moderate acute malnutrition (MAM) with persistent microbiota immaturity. Host and microbial effects of microbiota-directed complementary food (MDCF) prototypes targeting weaning-phase bacterial taxa underrepresented in SAM and MAM microbiota were characterized in gnotobiotic mice and gnotobiotic piglets colonized with age- and growth-discriminatory bacteria. A randomized, double-blind controlled feeding study identified a lead MDCF that changes the abundances of targeted bacteria and increases plasma biomarkers and mediators of growth, bone formation, neurodevelopment, and immune function in children with MAM.
Collapse
Affiliation(s)
- Jeanette L Gehrig
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Siddarth Venkatesh
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hao-Wei Chang
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Matthew C Hibberd
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Vanderlene L Kung
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jiye Cheng
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Robert Y Chen
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sathish Subramanian
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carrie A Cowardin
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Martin F Meier
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David O'Donnell
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael Talcott
- Division of Comparative Medicine, Washington University, St. Louis, MO 63110, USA
| | - Larry D Spears
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Clay F Semenkovich
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Bernard Henrissat
- Architecture et Fonction des Macromolécules Biologiques, Centre National de la Recherche Scientifique and Aix-Marseille Université, 13288 Marseille cedex 9, France
- Department of Biological Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Richard J Giannone
- Chemical Sciences Division, Oak Ridge National Laboratory, Oak Ridge, TN 37830, USA
| | - Robert L Hettich
- Chemical Sciences Division, Oak Ridge National Laboratory, Oak Ridge, TN 37830, USA
| | - Olga Ilkayeva
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27710, USA
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Michael Muehlbauer
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27710, USA
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27710, USA
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27710, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Christopher Sawyer
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Genome Technology Access Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Richard D Head
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Genome Technology Access Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Dmitry A Rodionov
- A. A. Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow 127994, Russia
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Aleksandr A Arzamasov
- A. A. Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow 127994, Russia
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Semen A Leyn
- A. A. Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow 127994, Russia
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Andrei L Osterman
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Md Iqbal Hossain
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh
| | - Munirul Islam
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh
| | - Nuzhat Choudhury
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh
| | - Shafiqul Alam Sarker
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh
| | - Sayeeda Huq
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh
| | - Imteaz Mahmud
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh
| | - Ishita Mostafa
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh
| | - Mustafa Mahfuz
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh
| | - Michael J Barratt
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tahmeed Ahmed
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh
| | - Jeffrey I Gordon
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA.
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
326
|
Figeac N, Mohamed AD, Sun C, Schönfelder M, Matallanas D, Garcia-Munoz A, Missiaglia E, Collie-Duguid E, De Mello V, Pobbati AV, Pruller J, Jaka O, Harridge SDR, Hong W, Shipley J, Vargesson N, Zammit PS, Wackerhage H. VGLL3 operates via TEAD1, TEAD3 and TEAD4 to influence myogenesis in skeletal muscle. J Cell Sci 2019; 132:jcs.225946. [PMID: 31138678 PMCID: PMC6633393 DOI: 10.1242/jcs.225946] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 05/03/2019] [Indexed: 12/21/2022] Open
Abstract
VGLL proteins are transcriptional co-factors that bind TEAD family transcription factors to regulate events ranging from wing development in fly, to muscle fibre composition and immune function in mice. Here, we characterise Vgll3 in skeletal muscle. We found that mouse Vgll3 was expressed at low levels in healthy muscle but that its levels increased during hypertrophy or regeneration; in humans, VGLL3 was highly expressed in tissues from patients with various muscle diseases, such as in dystrophic muscle and alveolar rhabdomyosarcoma. Interaction proteomics revealed that VGLL3 bound TEAD1, TEAD3 and TEAD4 in myoblasts and/or myotubes. However, there was no interaction with proteins from major regulatory systems such as the Hippo kinase cascade, unlike what is found for the TEAD co-factors YAP (encoded by YAP1) and TAZ (encoded by WWTR1). Vgll3 overexpression reduced the activity of the Hippo negative-feedback loop, affecting expression of muscle-regulating genes including Myf5, Pitx2 and Pitx3, and genes encoding certain Wnts and IGFBPs. VGLL3 mainly repressed gene expression, regulating similar genes to those regulated by YAP and TAZ. siRNA-mediated Vgll3 knockdown suppressed myoblast proliferation, whereas Vgll3 overexpression strongly promoted myogenic differentiation. However, skeletal muscle was overtly normal in Vgll3-null mice, presumably due to feedback signalling and/or redundancy. This work identifies VGLL3 as a transcriptional co-factor operating with the Hippo signal transduction network to control myogenesis. Summary: VGLL3 interacts with TEAD transcription factors to direct expression of crucial muscle regulatory genes and contribute to the control of skeletal myogenesis.
Collapse
Affiliation(s)
- Nicolas Figeac
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - Abdalla D Mohamed
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK.,Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environment and Health, Ingolstaedter Landstrasse 1, D-85764 Munich/Neuherberg, Germany
| | - Congshan Sun
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK.,Department of Neurology, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Martin Schönfelder
- Faculty of Sport and Health Sciences, Technical University of Munich, Georg-Brauchle-Ring 60, 80992 Munich, Germany
| | - David Matallanas
- Systems Biology Ireland, Conway Institute, Belfield; Dublin 4, Ireland
| | | | - Edoardo Missiaglia
- Institute of Pathology, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| | - Elaina Collie-Duguid
- University of Aberdeen, Centre for Genome Enabled Biology and Medicine, 23 St Machar Drive, Aberdeen AB24 3RY, UK
| | - Vanessa De Mello
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Ajaybabu V Pobbati
- Institute of Molecular and Cell Biology, A-STAR, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Johanna Pruller
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - Oihane Jaka
- Centre for Human and Applied Physiological Sciences, King's College London, London SE1 1UL, UK
| | - Stephen D R Harridge
- Centre for Human and Applied Physiological Sciences, King's College London, London SE1 1UL, UK
| | - Wanjin Hong
- Institute of Molecular and Cell Biology, A-STAR, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Janet Shipley
- Sarcoma Molecular Pathology Team, Divisions of Molecular Pathology and Cancer Therapeutics, Institute of Cancer Research, Surrey, SM2 5NG, UK
| | - Neil Vargesson
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Peter S Zammit
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - Henning Wackerhage
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK .,Faculty of Sport and Health Sciences, Technical University of Munich, Georg-Brauchle-Ring 60, 80992 Munich, Germany
| |
Collapse
|
327
|
Manzella L, Massimino M, Stella S, Tirrò E, Pennisi MS, Martorana F, Motta G, Vitale SR, Puma A, Romano C, Di Gregorio S, Russo M, Malandrino P, Vigneri P. Activation of the IGF Axis in Thyroid Cancer: Implications for Tumorigenesis and Treatment. Int J Mol Sci 2019; 20:E3258. [PMID: 31269742 PMCID: PMC6651760 DOI: 10.3390/ijms20133258] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 06/28/2019] [Accepted: 06/28/2019] [Indexed: 12/22/2022] Open
Abstract
The Insulin-like growth factor (IGF) axis is one of the best-established drivers of thyroid transformation, as thyroid cancer cells overexpress both IGF ligands and their receptors. Thyroid neoplasms encompass distinct clinical and biological entities as differentiated thyroid carcinomas (DTC)-comprising papillary (PTC) and follicular (FTC) tumors-respond to radioiodine therapy, while undifferentiated tumors-including poorly-differentiated (PDTC) or anaplastic thyroid carcinomas (ATCs)-are refractory to radioactive iodine and exhibit limited responses to chemotherapy. Thus, safe and effective treatments for the latter aggressive thyroid tumors are urgently needed. Despite a strong preclinical rationale for targeting the IGF axis in thyroid cancer, the results of the available clinical studies have been disappointing, possibly because of the crosstalk between IGF signaling and other pathways that may result in resistance to targeted agents aimed against individual components of these complex signaling networks. Based on these observations, the combinations between IGF-signaling inhibitors and other anti-tumor drugs, such as DNA damaging agents or kinase inhibitors, may represent a promising therapeutic strategy for undifferentiated thyroid carcinomas. In this review, we discuss the role of the IGF axis in thyroid tumorigenesis and also provide an update on the current knowledge of IGF-targeted combination therapies for thyroid cancer.
Collapse
Affiliation(s)
- Livia Manzella
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico-Vittorio Emanuele, 95123 Catania, Italy.
| | - Michele Massimino
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico-Vittorio Emanuele, 95123 Catania, Italy
| | - Stefania Stella
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico-Vittorio Emanuele, 95123 Catania, Italy
| | - Elena Tirrò
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico-Vittorio Emanuele, 95123 Catania, Italy
| | - Maria Stella Pennisi
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico-Vittorio Emanuele, 95123 Catania, Italy
| | - Federica Martorana
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico-Vittorio Emanuele, 95123 Catania, Italy
- Department of Medical Oncology A.O.U. Policlinico-Vittorio Emanuele, 95123 Catania, Italy
| | - Gianmarco Motta
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico-Vittorio Emanuele, 95123 Catania, Italy
- Department of Medical Oncology A.O.U. Policlinico-Vittorio Emanuele, 95123 Catania, Italy
| | - Silvia Rita Vitale
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico-Vittorio Emanuele, 95123 Catania, Italy
| | - Adriana Puma
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico-Vittorio Emanuele, 95123 Catania, Italy
| | - Chiara Romano
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico-Vittorio Emanuele, 95123 Catania, Italy
| | - Sandra Di Gregorio
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico-Vittorio Emanuele, 95123 Catania, Italy
| | - Marco Russo
- Endocrinology, Department of Clinical and Experimental Medicine, Garibaldi-Nesima Medical Center, University of Catania, 95122, Italy
| | - Pasqualino Malandrino
- Endocrinology, Department of Clinical and Experimental Medicine, Garibaldi-Nesima Medical Center, University of Catania, 95122, Italy
| | - Paolo Vigneri
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico-Vittorio Emanuele, 95123 Catania, Italy
| |
Collapse
|
328
|
Yan JJ, Hwang PP. Novel discoveries in acid-base regulation and osmoregulation: A review of selected hormonal actions in zebrafish and medaka. Gen Comp Endocrinol 2019; 277:20-29. [PMID: 30878350 DOI: 10.1016/j.ygcen.2019.03.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 02/26/2019] [Accepted: 03/12/2019] [Indexed: 12/28/2022]
Abstract
Maintenance of internal ionic and acid-base homeostasis is critical for survival in all biological systems. Similar to mammals, aquatic fishes have developed sophisticated homeostatic mechanisms to mitigate metabolic or environmental disruptions in ionic and acid-base status of systemic body fluids via hormone-controlled transport of ions or acid equivalents. The present review summarizes newly discovered actions of several hormones in zebrafish (Danio rerio) and medaka (Oryzias latipes) that have greatly contributed to our overall understanding of ionic/acid-base regulation. For example, isotocin and cortisol were reported to enhance transport of various ions by stimulating the proliferation and/or differentiation of ionocyte progenitors. Meanwhile, stanniocalcin-1, a well-documented hypocalcemic hormone, was found to suppress ionocyte differentiation and thus downregulate secretion of H+ and uptake of Na+ and Cl-. Estrogen-related receptor and calcitonin gene-related peptide also regulate the differentiation of certain types of ionocytes to either stimulate or suppress H+ secretion and Cl- uptake. On the other hand, endothelin and insulin-like growth factor 1 activate the respective secretion of H+ and Na+/Cl through fast actions. These new findings enhance our understanding of how hormones regulate fish ionic and acid-base regulation while further providing new insights into vertebrate evolution, mammalian endocrinology and human disease-related therapeutics.
Collapse
Affiliation(s)
- Jia-Jiun Yan
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Pung-Pung Hwang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
329
|
Czaja W, Nakamura YK, Li N, Eldridge JA, DeAvila DM, Thompson TB, Rodgers BD. Myostatin regulates pituitary development and hepatic IGF1. Am J Physiol Endocrinol Metab 2019; 316:E1036-E1049. [PMID: 30888862 PMCID: PMC6620572 DOI: 10.1152/ajpendo.00001.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Circulating myostatin-attenuating agents are being developed to treat muscle-wasting disease despite their potential to produce serious off-target effects, as myostatin/activin receptors are widely distributed among many nonmuscle tissues. Our studies suggest that the myokine not only inhibits striated muscle growth but also regulates pituitary development and growth hormone (GH) action in the liver. Using a novel myostatin-null label-retaining model (Jekyll mice), we determined that the heterogeneous pool of pituitary stem, transit-amplifying, and progenitor cells in Jekyll mice depletes more rapidly after birth than the pool in wild-type mice. This correlated with increased levels of GH, prolactin, and the cells that secrete these hormones, somatotropes and lactotropes, respectively, in Jekyll pituitaries. Recombinant myostatin also stimulated GH release and gene expression in pituitary cell cultures although inhibiting prolactin release. In primary hepatocytes, recombinant myostatin blocked GH-stimulated expression of two key mediators of growth, insulin-like growth factor (IGF)1 and the acid labile subunit and increased expression of an inhibitor, IGF-binding protein-1. The significance of these findings was demonstrated by smaller muscle fiber size in a model lacking myostatin and liver IGF1 expression (LID-o-Mighty mice) compared with that in myostatin-null (Mighty) mice. These data together suggest that myostatin may regulate pituitary development and function and that its inhibitory actions in muscle may be partly mediated by attenuating GH action in the liver. They also suggest that circulating pharmacological inhibitors of myostatin could produce unintended consequences in these and possibly other tissues.
Collapse
Affiliation(s)
- Wioletta Czaja
- Washington Center for Muscle Biology, Department of Animal Sciences, Washington State University , Pullman, Washington
- Department of Biochemistry and Molecular Biology, University of Georgia , Athens, Georgia
| | - Yukiko K Nakamura
- Washington Center for Muscle Biology, Department of Animal Sciences, Washington State University , Pullman, Washington
| | - Naisi Li
- Washington Center for Muscle Biology, Department of Animal Sciences, Washington State University , Pullman, Washington
| | - Jennifer A Eldridge
- Washington Center for Muscle Biology, Department of Animal Sciences, Washington State University , Pullman, Washington
| | - David M DeAvila
- Washington Center for Muscle Biology, Department of Animal Sciences, Washington State University , Pullman, Washington
| | - Thomas B Thompson
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati , Cincinnati, Ohio
| | - Buel D Rodgers
- Washington Center for Muscle Biology, Department of Animal Sciences, Washington State University , Pullman, Washington
- AAVogen, Incorporated, Rockville, Maryland
| |
Collapse
|
330
|
Higashi Y, Gautam S, Delafontaine P, Sukhanov S. IGF-1 and cardiovascular disease. Growth Horm IGF Res 2019; 45:6-16. [PMID: 30735831 PMCID: PMC6504961 DOI: 10.1016/j.ghir.2019.01.002] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 10/17/2018] [Accepted: 01/30/2019] [Indexed: 12/14/2022]
Abstract
Atherosclerosis is an inflammatory arterial pathogenic condition, which leads to ischemic cardiovascular diseases, such as coronary artery disease and myocardial infarction, stroke, and peripheral arterial disease. Atherosclerosis is a multifactorial disorder and its pathophysiology is highly complex. Changes in expression of multiple genes coupled with environmental and lifestyle factors initiate cascades of adverse events involving multiple types of cells (e.g. vascular endothelial cells, smooth muscle cells, and macrophages). IGF-1 is a pleiotropic factor, which is found in the circulation (endocrine IGF-1) and is also produced locally in arteries (endothelial cells and smooth muscle cells). IGF-1 exerts a variety of effects on these cell types in the context of the pathogenesis of atherosclerosis. In fact, there is an increasing body of evidence suggesting that IGF-1 has beneficial effects on the biology of atherosclerosis. This review will discuss recent findings relating to clinical investigations on the relation between IGF-1 and cardiovascular disease and basic research using animal models of atherosclerosis that have elucidated some of the mechanisms underlying atheroprotective effects of IGF-1.
Collapse
Affiliation(s)
- Yusuke Higashi
- Department of Medicine, School of Medicine, University of Missouri, Columbia, MO, United States; Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States.
| | - Sandeep Gautam
- Department of Medicine, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Patrick Delafontaine
- Department of Medicine, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Sergiy Sukhanov
- Department of Medicine, School of Medicine, University of Missouri, Columbia, MO, United States
| |
Collapse
|
331
|
Slater T, Haywood NJ, Matthews C, Cheema H, Wheatcroft SB. Insulin-like growth factor binding proteins and angiogenesis: from cancer to cardiovascular disease. Cytokine Growth Factor Rev 2019; 46:28-35. [PMID: 30954375 DOI: 10.1016/j.cytogfr.2019.03.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 03/15/2019] [Indexed: 12/22/2022]
Abstract
Angiogenesis is a tightly regulated activity that is vital during embryonic development and for normal physiological repair processes and reproduction in healthy adults. Pathological angiogenesis is a driving force behind a variety of diseases including cancer and retinopathies, and inhibition of angiogenesis is a therapeutic option that has been the subject of much research, with several inhibitory agents now available for medical therapy. Conversely, therapeutic angiogenesis has been mooted as having significant potential in the treatment of ischemic conditions such as angina pectoris and peripheral arterial disease, but so far there has been less translation from lab to bedside. The insulin-like growth factor binding proteins (IGFBP) are a family of seven proteins essential for the binding and transport of the insulin-like growth factors (IGF). It is being increasingly recognised that IGFBPs have a significant role beyond simply modulating IGF activity, with evidence of both IGF dependent and independent actions through a variety of mechanisms. Moreover, the action of the IGFBPs can be stimulatory or inhibitory depending on the cell type and environment. Specifically the IGFBPs have been heavily implicated in angiogenesis, both pathological and physiological, and they have significant promise as targeted cell therapy agents for both pathological angiogenesis inhibition and therapeutic angiogenesis following ischemic injury. In this short review we will explore the current understanding of the individual impact of each IGFBP on angiogenesis, and the pathways through which these effects occur.
Collapse
Affiliation(s)
- Thomas Slater
- Leeds Institute of Cardiovascular & Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom
| | - Natalie J Haywood
- Leeds Institute of Cardiovascular & Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom
| | - Connor Matthews
- Leeds Institute of Cardiovascular & Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom
| | - Harneet Cheema
- Leeds Institute of Cardiovascular & Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom
| | - Stephen B Wheatcroft
- Leeds Institute of Cardiovascular & Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom.
| |
Collapse
|
332
|
Cleveland BM, Yamaguchi G, Radler LM, Shimizu M. Editing the duplicated insulin-like growth factor binding protein-2b gene in rainbow trout (Oncorhynchus mykiss). Sci Rep 2018; 8:16054. [PMID: 30375441 PMCID: PMC6207780 DOI: 10.1038/s41598-018-34326-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 10/17/2018] [Indexed: 01/10/2023] Open
Abstract
In salmonids, the majority of circulating insulin-like growth factor-I (IGF-I) is bound to IGF binding proteins (IGFBP), with IGFBP-2b being the most abundant in circulation. We used CRISPR/Cas9 methodology to disrupt expression of a functional IGFBP-2b protein by co-targeting for gene editing IGFBP-2b1 and IGFBP-2b2 subtypes, which represent salmonid-specific gene duplicates. Twenty-four rainbow trout were produced with mutations in the IGFBP-2b1 and IGFBP-2b2 genes. Mutant fish exhibited between 8–100% and 2–83% gene disruption for IGFBP-2b1 and IGFBP-2b2, respectively, with a positive correlation (P < 0.001) in gene mutation rate between individual fish. Analysis of IGFBP-2b protein indicated reductions in plasma IGFBP-2b abundance to between 0.04–0.96-fold of control levels. Plasma IGF-I, body weight, and fork length were reduced in mutants at 8 and 10 months post-hatch, which supports that IGFBP-2b is significant for carrying IGF-I. Despite reduced plasma IGF-I and IGFBP-2b in mutants, growth retardation in mutants was less severe between 10 and 12 months post-hatch (P < 0.05), suggesting a compensatory growth response occurred. These findings indicate that gene editing using CRISPR/Cas9 and ligand blotting is a feasible approach for characterizing protein-level functions of duplicated IGFBP genes in salmonids and is useful to unravel IGF-related endocrine mechanisms.
Collapse
Affiliation(s)
- Beth M Cleveland
- National Center for Cool and Cold Water Aquaculture, Agricultural Research Service, United States Department of Agriculture, Kearneysville, West Virginia, United States of America.
| | - Ginnosuke Yamaguchi
- Graduate School of Environmental Science, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Lisa M Radler
- National Center for Cool and Cold Water Aquaculture, Agricultural Research Service, United States Department of Agriculture, Kearneysville, West Virginia, United States of America
| | - Munetaka Shimizu
- Faculty of Fisheries Sciences, Hokkaido University, Hakodate, Hokkaido, Japan
| |
Collapse
|
333
|
Liu C, Xin Y, Bai Y, Lewin G, He G, Mai K, Duan C. Ca 2+ concentration-dependent premature death of igfbp5a-/- fish reveals a critical role of IGF signaling in adaptive epithelial growth. Sci Signal 2018; 11:11/548/eaat2231. [PMID: 30228225 DOI: 10.1126/scisignal.aat2231] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The phenotype gap is a challenge for genetically dissecting redundant endocrine signaling pathways, such as the six isoforms in the insulin-like growth factor binding protein (IGFBP) family. Although overexpressed IGFBPs can inhibit or potentiate IGF actions or have IGF-independent actions, mutant mice lacking IGFBP-encoding genes do not exhibit major phenotypes. We found that although zebrafish deficient in igfbp5a did not show overt phenotypes when raised in Ca2+-rich solutions, they died prematurely in low Ca2+ conditions. A group of epithelial cells expressing igfbp5a take up Ca2+ and proliferate under low Ca2+ conditions because of activation of IGF signaling. Deletion of igfbp5a blunted low Ca2+ stress-induced IGF signaling and impaired adaptive proliferation. Reintroducing zebrafish Igfbp5a, but not its ligand binding-deficient mutant, restored adaptive proliferation. Similarly, adaptive proliferation was restored in zebrafish lacking igfbp5a by expression of human IGFBP5, but not two cancer-associated IGFBP5 mutants. Knockdown of IGFBP5 in human colon carcinoma cells resulted in reduced IGF-stimulated cell proliferation. These results reveal a conserved mechanism by which a locally expressed Igfbp regulates organismal Ca2+ homeostasis and survival by activating IGF signaling in epithelial cells and promoting their proliferation in Ca2+-deficient states. These findings underscore the importance of physiological context when analyzing loss-of-function phenotypes of endocrine factors.
Collapse
Affiliation(s)
- Chengdong Liu
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA.,The Key Laboratory of Mariculture, Education Ministry of China and College of Fisheries, Ocean University of China, Qingdao 266003, China
| | - Yi Xin
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yan Bai
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Grant Lewin
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Gen He
- The Key Laboratory of Mariculture, Education Ministry of China and College of Fisheries, Ocean University of China, Qingdao 266003, China.,Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, China
| | - Kangsen Mai
- The Key Laboratory of Mariculture, Education Ministry of China and College of Fisheries, Ocean University of China, Qingdao 266003, China.,Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, China
| | - Cunming Duan
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
334
|
Neurite Growth and Polarization on Vitronectin Substrate after in Vitro Trauma is not Enhanced after IGF Treatment. Brain Sci 2018; 8:brainsci8080151. [PMID: 30103517 PMCID: PMC6119911 DOI: 10.3390/brainsci8080151] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 07/31/2018] [Accepted: 08/08/2018] [Indexed: 11/23/2022] Open
Abstract
Following traumatic brain injuries (TBI), insulin-like growth factor (IGF) is cortically widely upregulated. This upregulation has a potential role in the recovery of neuronal tissue, plasticity, and neurotrophic activity, though the molecular mechanisms involved in IGF regulation and the exact role of IGF after TBI remain unclear. Vitronectin (VN), an extracellular matrix (ECM) molecule, has recently been shown to be of importance for IGF-mediated cellular growth and migration. Since VN is downregulated after TBI, we hypothesized that insufficient VN levels after TBI impairs the potential beneficial activity of IGF. To test if vitronectin and IGF-1/IGFBP-2 could contribute to neurite growth, we cultured hippocampal neurons on ± vitronectin-coated coverslips and them treated with ± IGF-1/IGF binding protein 2 (IGFBP-2). Under same conditions, cell cultures were also subjected to in vitro trauma to investigate differences in the posttraumatic regenerative capacity with ± vitronectin-coated coverslips and with ± IGF-1/IGFBP-2 treatment. In both the control and trauma situations, hippocampal neurons showed a stronger growth pattern on vitronectin than on the control substrate. Surprisingly, the addition of IGF-1/IGFBP-2 showed a decrease in neurite growth. Since neurite growth was measured as the number of neurites per area, we hypothesized that IGF-1/IGFBP-2 contributes to the polarization of neurons and thus induced a less dense neurite network after IGF-1/IGFBP-2 treatment. This hypothesis could not be confirmed and we therefore conclude that vitronectin has a positive effect on neurite growth in vitro both under normal conditions and after trauma, but that addition of IGF-1/IGFBP-2 does not have a positive additive effect.
Collapse
|
335
|
Steffensen LL, Ernst EH, Amoushahi M, Ernst E, Lykke-Hartmann K. Transcripts Encoding the Androgen Receptor and IGF-Related Molecules Are Differently Expressed in Human Granulosa Cells From Primordial and Primary Follicles. Front Cell Dev Biol 2018; 6:85. [PMID: 30148131 PMCID: PMC6095988 DOI: 10.3389/fcell.2018.00085] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 07/18/2018] [Indexed: 11/13/2022] Open
Abstract
Bidirectional cross talk between granulosa cells and oocytes is known to be important in all stages of mammalian follicular development. Insulin-like growth factor (IGF) signaling is a prominent candidate to be involved in the activation of primordial follicles, and may be be connected to androgen-signaling. In this study, we interrogated transcriptome dynamics in granulosa cells isolated from human primordial and primary follicles to reveal information of growth factors and androgens involved in the physiology of ovarian follicular activation. Toward this, a transcriptome comparison study on primordial follicles (n = 539 follicles) and primary follicles (n = 261 follicles) donated by three women having ovarian tissue cryopreserved before chemotherapy was performed. The granulosa cell contribution in whole follicle isolates was extracted in silico. Modeling of complex biological systems was performed using IPA® software. We found the granulosa cell compartment of the human primordial and primary follicles to be extensively enriched in genes encoding IGF-related factors, and the Androgen Receptor (AR) enriched in granulosa cells of primordial follicles. Our study hints the possibility that primordial follicles may indeed be androgen responsive, and that the action of androgens represents a connection to the expression of key players in the IGF-signaling pathway including IGF1R, IGF2, and IGFBP3, and that this interaction could be important for early follicular activation. In line with this, several androgen-responsive genes were noted to be expressed in both oocytes and granulosa cells from human primordial and primary follicle. We present a detailed description of AR and IGF gene activities in the human granulosa cell compartment of primordial and primary follicles, suggesting that these cells may be or prepare to be responsive toward androgens and IGFs.
Collapse
Affiliation(s)
| | - Emil H Ernst
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Erik Ernst
- The Fertility Clinic, Horsens Hospital, Horsens, Denmark.,The Fertility Clinic, Aarhus University Hospital, Aarhus, Denmark
| | - Karin Lykke-Hartmann
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Clinical Genetics, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
336
|
Hjortebjerg R. IGFBP-4 and PAPP-A in normal physiology and disease. Growth Horm IGF Res 2018; 41:7-22. [PMID: 29864720 DOI: 10.1016/j.ghir.2018.05.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 05/15/2018] [Accepted: 05/29/2018] [Indexed: 02/07/2023]
Abstract
Insulin-like growth factor (IGF) binding protein-4 (IGFBP-4) is a modulator of the IGF system, exerting both inhibitory and stimulatory effects on IGF-induced cellular growth. IGFBP-4 is the principal substrate for the enzyme pregnancy-associated plasma protein-A (PAPP-A). Through IGF-dependent cleavage of IGFBP-4 in the vicinity of the IGF receptor, PAPP-A is able to increase IGF bioavailability and stimulate IGF-mediated growth. Recently, the stanniocalcins (STCs) were identified as novel inhibitors of PAPP-A proteolytic activity, hereby adding additional members to the seemingly endless list of proteins belonging to the IGF family. Our understanding of these proteins has advanced throughout recent years, and there is evidence to suggest that the role of IGFBP-4 and PAPP-A in defining the relationship between total IGF and IGF bioactivity can be linked to a number of pathological conditions. This review provides an overview of the experimental and clinical findings on the IGFBP-4/PAPP-A/STC axis as a regulator of IGF activity and examines the conundrum surrounding extrapolation of circulating concentrations to tissue action of these proteins. The primary focus will be on the biological significance of IGFBP-4 and PAPP-A in normal physiology and in pathophysiology with emphasis on metabolic disorders, cardiovascular diseases, and cancer. Finally, the review assesses current new trajectories of IGFBP-4 and PAPP-A research.
Collapse
Affiliation(s)
- Rikke Hjortebjerg
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark; The Danish Diabetes Academy, Odense, Denmark.
| |
Collapse
|
337
|
Pérez-Sánchez J, Simó-Mirabet P, Naya-Català F, Martos-Sitcha JA, Perera E, Bermejo-Nogales A, Benedito-Palos L, Calduch-Giner JA. Somatotropic Axis Regulation Unravels the Differential Effects of Nutritional and Environmental Factors in Growth Performance of Marine Farmed Fishes. Front Endocrinol (Lausanne) 2018; 9:687. [PMID: 30538673 PMCID: PMC6277588 DOI: 10.3389/fendo.2018.00687] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 11/02/2018] [Indexed: 12/23/2022] Open
Abstract
The Gh/Prl/Sl family has evolved differentially through evolution, resulting in varying relationships between the somatotropic axis and growth rates within and across fish species. This is due to a wide range of endogenous and exogenous factors that make this association variable throughout season and life cycle, and the present minireview aims to better define the nutritional and environmental regulation of the endocrine growth cascade over precisely defined groups of fishes, focusing on Mediterranean farmed fishes. As a result, circulating Gh and Igf-i are revitalized as reliable growth markers, with a close association with growth rates of gilthead sea bream juveniles with deficiency signs in both macro- or micro-nutrients. This, together with other regulated responses, promotes the use of Gh and Igf-i as key performance indicators of growth, aerobic scope, and nutritional condition in gilthead sea bream. Moreover, the sirtuin-energy sensors might modulate the growth-promoting action of somatotropic axis. In this scenario, transcripts of igf-i and gh receptors mirror changes in plasma Gh and Igf-i levels, with the ghr-i/ghr-ii expression ratio mostly unaltered over season. However, this ratio is nutritionally regulated, and enriched plant-based diets or diets with specific nutrient deficiencies downregulate hepatic ghr-i, decreasing the ghr-i/ghr-ii ratio. The same trend, due to a ghr-ii increase, is found in skeletal muscle, whereas impaired growth during overwintering is related to increase in the ghr-i/ghr-ii and igf-ii/igf-i ratios in liver and skeletal muscle, respectively. Overall, expression of insulin receptors and igf receptors is less regulated, though the expression quotient is especially high in the liver and muscle of sea bream. Nutritional and environmental regulation of the full Igf binding protein 1-6 repertoire remains to be understood. However, tissue-specific expression profiling highlights an enhanced and nutritionally regulated expression of the igfbp-1/-2/-4 clade in liver, whereas the igfbp-3/-5/-6 clade is overexpressed and regulated in skeletal muscle. The somatotropic axis is, therefore, highly informative of a wide-range of growth-disturbing and stressful stimuli, and multivariate analysis supports its use as a reliable toolset for the assessment of growth potentiality and nutrient deficiencies and requirements, especially in combination with selected panels of other nutritionally regulated metabolic biomarkers.
Collapse
|