Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
2.
Article in English | MEDLINE | ID: mdl-28899845

ABSTRACT

Growth is one of the main biological processes in aquatic organisms that is affected by environmental fluctuations such as upwelling (characterized by food-rich waters). In fish, growth is directly related with skeletal muscle increase; which represents the largest tissue of body mass. However, the effects of upwelling on growth, at the physiological and molecular level, are unknown. This study used Girella laevifrons (one of the most abundant intertidal fish in Eastern South Pacific) as a biological model, considering animals from upwelling (U) and non-upwelling (NU) areas. Here, we evaluated the effect of nutritional composition and food availability on growth performance and expression of key growth-related genes (insulin-kike growth factor 1 (igf1) and myosin heavy-chain (myhc)) and atrophy-related genes (muscle ring-finger 1 (murf1), F-box only protein 32 (atrogin-1) and BCL2/adenovirus E1B 19kDa-interacting protein 3 (bnip3)). We reported that, among zones, U fish displayed higher growth performance in response to nutritional composition, specifically between protein- and fiber-rich diets (~1g). We also found in NU fish that atrophy-related genes were upregulated with fiber-rich diet and during fasting (~2-fold at minimum respect U). In conclusion, our results suggest that the growth potential of upwelling fish may be a consequence of differential muscle gene expression. Our data provide a preliminary approach contributing on how upwelling influence fish growth at the physiological and molecular levels. Future studies are required to gain further knowledge about molecular differences between U and NU animals, as well as the possible applications of this knowledge in the aquaculture industry.


Subject(s)
Gene Expression Regulation, Developmental , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Perciformes/growth & development , Perciformes/genetics , Animals , Diet, Carbohydrate Loading , Ecosystem , Food Chain , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/growth & development , Perciformes/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Rivers/chemistry , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Seawater/chemistry , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism
3.
Gen Comp Endocrinol ; 247: 53-65, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28109823

ABSTRACT

Much attention has been given to insulin-like growth factor (Igf) pathways that regulate the balance of skeletal muscle protein synthesis and breakdown in response to a range of extrinsic and intrinsic signals. However, we have a less complete understanding of how the same signals modulate muscle mass upstream of such signalling, through a family of functionally-diverse Igf-binding proteins (Igfbps) that modify the availability of Igfs to the cell receptor Igf1r. We exposed cultured myotubes from Atlantic salmon (Salmo salar L.) to treatments recapturing three catabolic signals: inflammation (interleukin-1ß), stress (dexamethasone) and fasting (amino acid deprivation), plus one anabolic signal: recovery of muscle mass post-fasting (supplementation of fasted myotubes with Igf-I and amino acids). The intended phenotype of treatments was confirmed by significant changes in myotube diameter and immunofluorescent staining of structural proteins. We quantified the mRNA-level regulation of the full expressed Igf and Igfbp gene complement across a post-treatment time course, along with marker genes for muscle structural protein synthesis, as well as muscle breakdown, via the ubiquitin-proteasome and autophagy systems. Our results highlight complex, non-overlapping responses of Igfbp family members to the different treatments, suggesting that the profile of expressed Igfbps is differentially regulated by distinct signals promoting similar muscle remodelling phenotypes. We also demonstrate divergent regulation of salmonid-specific gene duplicates of igfbp5b1 and igfbp5b2 under distinct catabolic and anabolic conditions. Overall, this study increases our understanding of the regulation of Igfbp genes in response to signals that promote remodelling of skeletal muscle.


Subject(s)
Gene Expression Regulation , Insulin-Like Growth Factor Binding Proteins/genetics , Muscle Fibers, Skeletal/metabolism , Salmo salar/genetics , Salmo salar/metabolism , Amino Acids/deficiency , Animals , Cells, Cultured , Dexamethasone/pharmacology , Gene Expression Regulation/drug effects , Humans , Insulin-Like Growth Factor Binding Proteins/metabolism , Insulin-Like Growth Factor I/pharmacology , Interleukin-1beta/pharmacology , Linear Models , Muscle Fibers, Skeletal/drug effects , Muscle Proteins/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects
4.
Gen Comp Endocrinol ; 210: 96-106, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25449137

ABSTRACT

Knowledge about the underlying mechanisms, particularly the signaling pathways that account for muscle growth in vivo in early vertebrates is still scarce. Fish (Paralichthys adspersus) were fasted for 3weeks to induce a catabolic period of strong muscle atrophy. Subsequently, fish were refed for 2weeks to induce compensatory muscle hypertrophy. During refeeding, the fish were treated daily with either rapamycin (TORC blocker), PD98059 (MEK blocker), or PBS (V; vehicle), or were untreated (C; control). Rapamycin and PD98059 differentially impaired muscle cellularity in vivo, growth performance, and the expression of growth-related genes, and the inhibition of TORC1 had a greater impact on fish muscle growth than the inhibition of MAPK. Blocking TORC1 inhibited the phosphorylation of P70S6K and 4EBP1, two downstream components activated by TORC1, thus affecting protein contents in muscle. Concomitantly, the gene expression in muscle of igf-1, 2 and igfbp-4, 5 was down-regulated while the expression of atrogin-1, murf-1, and igfbp-2, 3 was up-regulated. Muscle hypertrophy was abolished and muscle atrophy was promoted, which finally affected body weight. TORC2 complex was not affected by rapamycin. On the other hand, the PD98059 treatment triggered ERK inactivation, a downstream component activated by MEK. mRNA contents of igf-1 in muscle were down-regulated, and muscle hypertrophy was partially impaired. The present study provides the first direct data on the in vivo contribution of TORC1/P70S6K, TORC1/4EBP1, and MAPK/ERK signaling pathways in the skeletal muscle of an earlier vertebrate, and highlights the transcendental role of TORC1 in growth from the cellular to organism level.


Subject(s)
Eukaryotic Initiation Factors/physiology , Flatfishes/growth & development , Mitogen-Activated Protein Kinase Kinases/physiology , Multiprotein Complexes/physiology , Muscle Development/physiology , Muscle, Skeletal/growth & development , Ribosomal Protein S6 Kinases, 70-kDa/physiology , Somatomedins/physiology , TOR Serine-Threonine Kinases/physiology , Animals , Flatfishes/metabolism , Flavonoids/pharmacology , Mechanistic Target of Rapamycin Complex 1 , Muscle Development/drug effects , Muscle Development/genetics , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Sirolimus/pharmacology
5.
FEBS Lett ; 588(23): 4390-7, 2014 Nov 28.
Article in English | MEDLINE | ID: mdl-25448676

ABSTRACT

Muscle-specific RING finger (MuRF) proteins are E3-ubiquitin ligases and key regulators of muscle growth and turnover. Here, using a range of phylogenomic approaches, we established the complete-definitive MuRF family of vertebrates. Adding to recognized MuRF1, 2 and 3, we describe a novel family member, hereafter MuRF4, which was independently lost during placental mammal and bird evolution, but is otherwise conserved. MuRF4 transcripts were expressed in heart and skeletal muscles of zebrafish, but were barely detectable in striated muscles of adult anole lizards. We also demonstrate that MuRF1 underwent retrotransposition in the teleost fish ancestor, before the retrogene fully replaced the original gene and muscle-specific function.


Subject(s)
Conserved Sequence , RING Finger Domains , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Vertebrates , Amino Acid Sequence , Animals , Evolution, Molecular , Gene Duplication , Gene Expression Regulation, Enzymologic , Genomics , Humans , Molecular Sequence Data , Phylogeny , Ubiquitin-Protein Ligases/genetics , Vertebrates/genetics
6.
Article in English | MEDLINE | ID: mdl-25088252

ABSTRACT

One of the most fundamental biological processes in living organisms that are affected by environmental fluctuations is growth. In fish, skeletal muscle accounts for the largest proportion of body mass, and the growth of this tissue is mainly controlled by the insulin-like growth factor (IGF) system. By using the carp (Cyprinus carpio), a fish that inhabits extreme conditions during winter and summer, we assessed the skeletal muscle plasticity induced by seasonal acclimatization and the relation of IGF signaling with protein synthesis and ribosomal biogenesis. The expression of igf1 in muscle decreased during winter in comparison with summer, whereas the expression for both paralogues of igf2 did not change significantly between seasons. The expression of igf1 receptor a (igf1ra), but not of igf1rb, was down-regulated in muscle during the winter as compared to the summer. A decrease in protein contents and protein phosphorylation for IGF signaling molecules in muscle was observed in winter-acclimatized carp. This was related with a decreased expression in muscle for markers of myogenesis (myoblast determination factor (myod), myogenic factor 5 (myf5), and myogenin (myog)); protein synthesis (myosin heavy chain (mhc) and myosin light chain (mlc3 and mlc1b)); and ribosomal biogenesis (pre-rRNA and ribosomal proteins). IGF signaling, and key markers of ribosomal biogenesis, protein synthesis, and myogenesis were affected by seasonal acclimatization, with differential regulation in gene expression and signaling pathway activation observed in muscle between both seasons. This suggests that these molecules are responsible for the muscle plasticity induced by seasonal acclimatization in carp.


Subject(s)
Acclimatization , Carps/metabolism , Insulin-Like Growth Factor I/metabolism , Muscle, Skeletal/metabolism , Ribosomes/metabolism , Animals , Biomarkers , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor II/metabolism , Ligands , Muscle Development , Muscle, Skeletal/growth & development , Phosphorylation , Protein Biosynthesis , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Ribosomes/genetics , Seasons , Signal Transduction
7.
Article in English | MEDLINE | ID: mdl-24769445

ABSTRACT

Ribosomal biogenesis controls cellular growth in living organisms, with the rate-limiting step of this process being the transcription of ribosomal DNA (rDNA). Considering that epigenetic mechanisms allow an organism to respond to environmental changes, the expression in muscle of several molecules that regulate epigenetic rRNA synthesis, as well as rDNA transcription, were evaluated during the seasonal acclimatization of the carp. First, the nucleotide sequences encoding the components forming the NoRC (ttf-I, tip5) and eNoSC (sirt1, nml, suv39h1), two chromatin remodeling complexes that silence rRNA synthesis, as well as the sequence of ubf1, a key regulator of rDNA transcription, were obtained. Subsequently the transcriptional regulation of the aforementioned molecules, and other key molecules involved in rRNA synthesis (mh2a1, mh2a2, h2a.z, h2a.z.7, nuc, p80), was assessed. The carp sequences for TTF-I, TIP5, SIRT1, NML, SUV39H1, and UBF1 showed a high conservation of domains and key amino acids in comparison with other fish and higher vertebrates. The mRNA contents in muscle for ttf-I, tip5, sirt1, nml, suv39h1, mh2a1, mh2a.z, and nuc were up-regulated during winter in comparison with summer, whereas the mRNA levels of mh2a2, ubf1, and p80 were down-regulated. Also, the contents of molecules involved in processing the rRNA (snoRNAs) and pRNA, a stabilizer of NoRC complex, were analyzed, finding that these non-coding RNAs were not affected by seasonal acclimatization. These results suggest that variations in the expression of rRNA and the molecules that epigenetically regulate its synthesis are contributing to the muscle plasticity induced by seasonal acclimatization in carp.


Subject(s)
Acclimatization , Carps/physiology , Muscle, Skeletal/physiology , RNA, Ribosomal/metabolism , Acclimatization/physiology , Animals , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone , Cloning, Molecular , Epigenesis, Genetic , Fish Proteins/genetics , Fish Proteins/metabolism , Gene Expression Regulation , Histones/genetics , Histones/metabolism , Pol1 Transcription Initiation Complex Proteins/genetics , Seasons , Sirtuin 1/genetics
8.
J Biotechnol ; 168(4): 295-302, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24184273

ABSTRACT

Myostatin (MSTN) is the main negative regulator of muscle growth and development in vertebrates. In fish, little is known about the molecular mechanisms behind how MSTN inactivation triggers skeletal muscle enhancement, particularly regarding the signaling pathways involved in this process. Moreover, there have not been reports on the biotechnological applications of MSTN and its signal transduction. In this context, zebrafish underwent compensatory growth using fasting and refeeding trials, and MSTN activity was inactivated with dominant negative LAPD76A recombinant proteins during the refeeding period, when a rapid, compensatory muscle growth was observed. Treated fish displayed an overcompensation of growth characterized by higher muscle hypertrophy and growth performance than constantly fed, control fish. Treatment with LAPD76A recombinant proteins triggered inactivation of the SMAD signaling pathway in skeletal muscle, the main signal transduction used by MSTN to achieve its biological actions. Therefore, transient inactivation of MSTN during the compensatory growth of zebrafish led to a decrease in the SMAD signaling pathway in muscle, triggering muscle hypertrophy and finally improving growth performance, thus, zebrafish achieved an overcompensation of growth. The present study shows an attractive strategy for improving muscle growth in a fish species by mixing a classical strategy, such as compensatory growth, and a biotechnological approach, such as the use of recombinant proteins for inhibiting the biological actions of MSTN. The mix of both strategies may represent a method that could be applied in order to improve growth in commercial fish of interest for aquaculture.


Subject(s)
Muscle, Skeletal/growth & development , Myostatin/genetics , Smad Proteins/genetics , Animals , Humans , Hypertrophy/genetics , Hypertrophy/metabolism , Muscle, Skeletal/metabolism , Myostatin/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Signal Transduction , Smad Proteins/antagonists & inhibitors , Zebrafish/genetics , Zebrafish/growth & development
9.
Gen Comp Endocrinol ; 192: 136-48, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23791761

ABSTRACT

The growth hormone (GH)-insulin-like growth factor (IGF) system is the key promoter of growth in vertebrates; however, how this system modulates muscle mass in fish is just recently becoming elucidated. In fish, the GH induces muscle growth by modulating the expression of several genes belonging to the myostatin (MSTN), atrophy, GH, and IGF systems as well as myogenic regulatory factors (MRFs). The GH controls the expression of igf1 via Janus kinase 2 (JAK2)/signal transducers and activators of the transcription 5 (STAT5) signaling pathway, but it seems that it is not the major regulator. These mild effects of the GH on igf1 expression in fish muscle seem to be related with the presence of higher contents of truncated GH receptor1 (tGHR1) than full length GHR (flGHR1). IGFs in fish stimulate myogenic cell proliferation, differentiation, and protein synthesis through the MAPK/ERK and PI3K/AKT/TOR signaling pathways, concomitant with abolishing protein degradation and atrophy via the PI3K/AKT/FOXO signaling pathway. Besides these signaling pathways control the expression of several genes belonging to the atrophy and IGF systems. Particularly, IGFs and amino acid control the expression of igf1, thus, suggesting other of alternative signaling pathways regulating the transcription of this growth factor. The possible role of IGF binding proteins (IGFBPs) and the contribution of muscle-derived versus hepatic-produced IGF1 on fish muscle growth is also addressed. Thus, a comprehensive overview on the GH-IGF system regulating fish skeletal muscle growth is presented, as well as perspectives for future research in this field.


Subject(s)
Growth Hormone/metabolism , Muscle, Skeletal/metabolism , Somatomedins/metabolism , Animals , Signal Transduction/physiology
10.
Gen Comp Endocrinol ; 186: 172-80, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23500005

ABSTRACT

Insight of how growth and metabolism in skeletal muscle are related is still lacking in early vertebrates. In this context, molecules involved in these processes, such as leptin, AMP-activated protein kinase (AMPK), target of rapamicyn (TOR), peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α, and oxidative phosphorylation complexes (OXPHOS), were assessed in the skeletal muscle of a fish species. Periods of fasting followed by a period of refeeding were implemented, using the fine flounder as a model (Paralichthys adspersus). This species exhibits remarkably slow growth and food intake, which is linked to an inherent growth hormone (GH) resistance and high circulating levels of leptin. Leptin increased during fasting concomitantly with AMPK activation, which was inversely correlated with TOR activation. On the other hand, AMPK was directly correlated with an increase in PGC-1α and OXPHOS complexes contents. Dramatic changes in the activation and content of these molecules were observed during short-term refeeding. Leptin, AMPK activation, and PGC-1α/OXPHOS complexes contents decreased radically; whereas, TOR activation increased significantly. During long-term refeeding these molecules returned to basal levels. These results suggest that there is a relation among these components; thus, during fasting periods ATP-consuming biosynthetic pathways are repressed and alternative sources of ATP/energy are promoted, a phenomenon that is reversed during anabolic periods. These results provide novel insight on the control of metabolism and growth in the skeletal muscle of a non-mammalian species, suggesting that both processes in fish muscle are closely related and coordinated by a subset of common molecules.


Subject(s)
AMP-Activated Protein Kinases/blood , Leptin/blood , Mitochondrial Turnover/physiology , Muscle, Skeletal/metabolism , Nutritional Status/physiology , Animals , Flounder/blood , Flounder/metabolism , PPAR gamma , Signal Transduction
11.
J Cell Physiol ; 228(7): 1452-63, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23255067

ABSTRACT

Skeletal muscle differentiation is a complex and highly regulated process characterized by cell cycle arrest, which is associated with morphological changes including myoblast alignment, elongation, and fusion into multinucleated myotubes. This is a balanced process dynamically coordinated by positive and negative signals such as the insulin-like growth factor I (IGF-1) and myostatin (MSTN), respectively. In this study, we report that the stimulation of skeletal myoblasts during differentiation with IGF-1 induces a rapid and transient calcium increase from intracellular stores, which are principally mediated through the phospholipase C gamma (PLC γ)/inositol 1,4,5-triphosphate (IP3 )-dependent signaling pathways. This response was completely blocked when myoblasts were incubated with LY294002 or transfected with the dominant-negative p110 gamma, suggesting a fundamental role of phosphatidylinositol 3-kinase (PI3K) in PLCγ activation. Additionally, we show that calcium released via IP3 and induced by IGF-1 stimulates NFAT-dependent gene transcription and nuclear translocation of the GFP-labeled NFATc3 isoform. This activation was independent of extracellular calcium influx and calcium release mediated by ryanodine receptor (RyR). Finally, we examined mstn mRNA levels and mstn promoter activity in myoblasts stimulated with IGF-1. We found a significant increase in mRNA contents and in reporter activity, which was inhibited by cyclosporin A, 11R-VIVIT, and by inhibitors of the PI3Kγ, PLCγ, and IP3 receptor. Our results strongly suggest that IGF-1 regulates myostatin transcription through the activation of the NFAT transcription factor in an IP3 /calcium-dependent manner. This is the first study to demonstrate a role of calcium-dependent signaling pathways in the mRNA expression of myostatin.


Subject(s)
Calcium Signaling/physiology , Inositol 1,4,5-Trisphosphate/metabolism , Insulin-Like Growth Factor I/metabolism , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/metabolism , Myostatin/genetics , NFATC Transcription Factors/metabolism , Animals , Calcium Signaling/genetics , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Gene Expression Regulation, Developmental , Phosphatidylinositol 3-Kinases/metabolism , Phospholipase C gamma/metabolism , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats
12.
Fish Physiol Biochem ; 39(4): 765-77, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23086610

ABSTRACT

In the present study, different reference genes were isolated, and their stability in the skeletal muscle of fine flounder subjected to different nutritional states was assessed using geNorm and NormFinder. The combinations between 18S and ActB; Fau and 18S; and Fau and Tubb were chosen as the most stable gene combinations in feeding, long-term fasting and refeeding, and short-term refeeding conditions, respectively. In all periods, ActB was identified as the single least stable gene. Subsequently, the expression of the myosin heavy chain (MYH) and the insulin-like growth factor-I receptor (IGF-IR) was assessed. A large variation in MYH and IGF-IR expression was found depending on the reference gene that was chosen for normalizing the expression of both genes. Using the most stable reference genes, mRNA levels of MYH decreased and IGF-IR increased during fasting, with both returning to basal levels during refeeding. However, the drop in mRNA levels for IGF-IR occurred during short-term refeeding, in contrast with the observed events in the expression of MYH, which occurred during long-term refeeding. The present study highlights the vast differences incurred when using unsuitable versus suitable reference genes for normalizing gene expression, pointing out that normalization without proper validation could result in a bias of gene expression.


Subject(s)
Flounder/genetics , Muscle, Skeletal/metabolism , Nutritional Status , Animals , Flounder/growth & development , Flounder/metabolism , Gene Expression , Myosin Heavy Chains/biosynthesis , Myosin Heavy Chains/genetics , Real-Time Polymerase Chain Reaction , Receptor, IGF Type 1/biosynthesis , Receptor, IGF Type 1/genetics
13.
PLoS One ; 7(9): e44256, 2012.
Article in English | MEDLINE | ID: mdl-23024748

ABSTRACT

A description of the intracellular mechanisms that modulate skeletal muscle atrophy in early vertebrates is still lacking. In this context, we used the fine flounder, a unique and intriguing fish model, which exhibits remarkably slow growth due to low production of muscle-derived IGF-I, a key growth factor that has been widely acknowledged to prevent and revert muscle atrophy. Key components of the atrophy system were examined in this species using a detailed time-course of sampling points, including two contrasting nutritional periods. Under basal conditions high amounts of the atrogenes MuRF-1 and Atrogin-1 were observed. During fasting, the activation of the P38/MAPK and Akt/FoxO signaling pathways decreased; whereas, the activation of the IκBα/NFκB pathway increased. These changes in signal transduction activation were concomitant with a strong increase in MuRF-1, Atrogin-1, and protein ubiquitination. During short-term refeeding, the P38/MAPK and Akt/FoxO signaling pathways were strongly activated, whereas the activation of the IκBα/NFκB pathway decreased significantly. The expression of both atrogenes, as well as the ubiquitination of proteins, dropped significantly during the first hour of refeeding, indicating a strong anti-atrophic condition during the onset of refeeding. During long-term refeeding, Akt remained activated at higher than basal levels until the end of refeeding, and Atrogin-1 expression remained significantly lower during this period. This study shows that the components of the atrophy system in skeletal muscle appeared early in the evolution of vertebrates and some mechanisms have been conserved, whereas others have not. These results represent an important achievement for the area of fish muscle physiology, showing an integrative view of the atrophy system in a non-mammalian species and contributing to novel insights on the molecular basis of muscle growth regulation in earlier vertebrates.


Subject(s)
Flounder/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Nutritional Status , Signal Transduction , Ubiquitinated Proteins/metabolism , Animals , Catalysis , Cluster Analysis , Female , Gene Expression Profiling , Gene Expression Regulation , Male , Muscle Proteins/genetics , Muscle, Skeletal/pathology , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Transcription, Genetic , Ubiquitinated Proteins/genetics , Ubiquitination
14.
J Endocrinol ; 214(1): 95-108, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22499735

ABSTRACT

The IGF-binding proteins (IGFBPs) play a dual role in the regulation of the activity and bioavailability of IGFs in different tissues. Diverse evidence has shown that IGFBPs can inhibit and/or potentiate IGF actions. In this study, igfbp1, 2, 3, 4, 5, and 6 were isolated in the fine flounder, a flat fish species that shows slow growth and inherent Gh resistance in muscle. Subsequently, the expression of all igfbps was assessed in the skeletal muscle of flounder that underwent different nutritional statuses. igfbp1 was not expressed in muscle during any of the nutritional conditions, whereas igfbp3 and igfbp5 were the lowest and the highest igfbps expressed respectively. A dynamic expression pattern was found in all the igfbps expressed in skeletal muscle, which depended on the nutritional status and sampling period. During the fasting period, igfbp2, 4, and 5 were downregulated, whereas igfbp3 was upregulated during part of the fasting period. The restoration of food modulated the expression of the igfbps dynamically, showing significant changes during both the long- and short-term refeeding. igfbp3 and igfbp6 were downregulated during short-term refeeding, whereas igfbp5 was upregulated, and igfbp2 and igfbp4 remained stable. During long-term refeeding, the expression of igfbp2, 4, 5, and 6 increased, while igfbp3 remained unchanged. In conclusion, this study shows for the first time the isolation of all igfbps in a single fish species, in addition to describing a dynamic nutritional and time-dependent response in the expression of igfbps in the skeletal muscle of a nonmammalian species.


Subject(s)
Animal Nutritional Physiological Phenomena , Fish Proteins/genetics , Flounder/genetics , Gene Expression Profiling , Insulin-Like Growth Factor Binding Proteins/genetics , Muscle, Skeletal/metabolism , Amino Acid Sequence , Animals , Autocrine Communication/genetics , Cluster Analysis , Eating/physiology , Fasting/physiology , Fish Proteins/classification , Insulin-Like Growth Factor Binding Protein 1/genetics , Insulin-Like Growth Factor Binding Protein 2/genetics , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 4/genetics , Insulin-Like Growth Factor Binding Protein 5/genetics , Insulin-Like Growth Factor Binding Protein 6/genetics , Insulin-Like Growth Factor Binding Proteins/classification , Molecular Sequence Data , Paracrine Communication/genetics , Phylogeny , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Time Factors
15.
Gen Comp Endocrinol ; 177(1): 120-7, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22429729

ABSTRACT

In fish, recent studies have indicated an anorexigenic role of leptin and thus its possible involvement in regulation of energy balance and growth. In the present study, the effects of fasting and refeeding periods on plasma leptin levels were studied in the fine flounder, a flatfish with remarkably slow growth. To further assess the endocrine status of the fish during periods of catabolism and anabolism, plasma growth hormone (GH) levels were also analyzed. Under normal feeding condition, plasma leptin and GH levels remained stable and relatively high in comparison with other teleost species. For the three separate groups of fish, fasted for 2, 3, and 4 weeks, respectively, plasma leptin levels increase gradually, becoming significantly elevated after 3 weeks, and reaching highest levels after 4-week fasting. Plasma GH levels were significantly elevated after 2-week fasting. At the onset of refeeding, following a single meal, leptin levels decline rapidly to lower than initial levels within 2 h, irrespective of the length of fasting. Plasma GH also decline, the decrease being significant after 4, 24 and 2 h for the 2, 3 and 4-week fasted groups, respectively. This study shows that plasma leptin levels in the fine flounder are strongly linked to nutritional status and suggests that leptin secretion is regulated by fast-acting mechanisms. Elevated leptin levels in fasted fish may contribute to a passive survival strategy of species which experience natural food shortage periods by lowering appetite and limiting physical foraging activity.


Subject(s)
Fasting/blood , Flounder/blood , Growth Hormone/blood , Leptin/blood , Animals , Postprandial Period/physiology
16.
Endocrinology ; 153(1): 283-94, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22028448

ABSTRACT

A detailed understanding of how the GH and IGF-I regulate muscle growth, especially in early vertebrates, is still lacking. The fine flounder is a flatfish species exhibiting remarkably slow growth, representing an intriguing model for elucidating growth regulatory mechanisms. Key components of the GH system were examined in groups of fish during periods of feeding, fasting, and refeeding. Under feeding conditions, there is an inherent systemic and local (muscle) GH resistance, characterized by higher levels of plasma GH than of IGF-I, skeletal muscle with a greater content of the truncated GH receptor (GHRt) than of full-length GHR (GHRfl), an impaired activation of the Janus kinase 2 (JAK2)-signal transducers and activators of transcription 5 (STAT5) signaling pathway, and low IGF-I expression. Fasting leads to further elevation of plasma GH levels concomitant with suppressed IGF-I levels. The ratio of GHRfl to GHRt in muscle decreases during fasting, causing an inactivation of the JAK2/STAT5 signaling pathway and suppressed IGF-I expression, further impairing growth. When fish are returned to nutritionally favorable conditions, plasma GH levels decrease, and the ratio of GHRfl to GHRt in muscle increases, triggering JAK2/STAT5 reactivation and local IGF-I expression, concomitant with increased growth. The study suggests that systemic IGF-I is supporting basal slow growth in this species, without ruling out that local IGF-I is participating in muscle growth. These results reveal for the first time a unique model of inherent GH resistance in the skeletal muscle of a nonmammalian species and contribute to novel insights of the endocrine and molecular basis of growth regulation in earlier vertebrates.


Subject(s)
Fish Proteins/metabolism , Flounder/metabolism , Growth Hormone/metabolism , Insulin-Like Growth Factor I/metabolism , Receptors, Somatotropin/metabolism , Animals , Base Sequence , Flounder/genetics , Flounder/growth & development , Growth Hormone/blood , Insulin-Like Growth Factor I/genetics , Janus Kinase 2/metabolism , Models, Biological , Muscle, Skeletal/metabolism , Nutritional Status , Peptide Fragments/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction
17.
Am J Physiol Regul Integr Comp Physiol ; 300(6): R1532-42, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21389330

ABSTRACT

The insulin-like growth factor-I (IGF-I) is a key regulator of skeletal muscle growth in vertebrates, promoting mitogenic and anabolic effects through the activation of the MAPK/ERK and the PI3K/Akt signaling pathways. Nutrition also affects skeletal muscle growth, activating intracellular pathways and inducing protein synthesis and accretion. Thus, both hormonal and nutritional signaling regulate muscle mass. In this context, plasma IGF-I levels and the activation of both pathways in response to food were evaluated in the fine flounder using fasting and refeeding trials. The present study describes for the first time in a nonmammalian species that the MAPK/ERK and PI3K/Akt are activated by exogenous circulating IGF-I, as well as showing that the MAPK/ERK pathway activation is modulated by the nutritional status. Also, these results show that there is a time-dependent regulation of IGF-I plasma levels and its signaling pathways in muscle. Together, these results suggest that the nutritionally managed IGF-I could be regulating the activation of the MAPK/ERK and the PI3K/Akt signaling pathways differentially according to the nutritional status, triggering different effects in growth parameters and therefore contributing to somatic growth in fish. This study contributes to the understanding of the nutrient regulation of IGF-I and its signaling pathways in skeletal muscle growth in nonmammalian species, therefore providing insight concerning the events controlling somatic growth in vertebrates.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/physiology , Flounder/growth & development , Flounder/physiology , Insulin-Like Growth Factor I/physiology , Mitogen-Activated Protein Kinase Kinases/physiology , Muscle, Skeletal/physiology , Nutritional Status/physiology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Animals , Eating/physiology , Fasting/physiology , Signal Transduction/physiology , Time Factors
18.
Article in English | MEDLINE | ID: mdl-21447399

ABSTRACT

Insulin-like growth factor-1 and insulin-like growth factor-1 receptor (IGF-1 and IGF-1R) play main roles in vertebrate growth and development. In fish, besides contributing to somatic growth, both molecules exhibit pleiotropic functions. We isolated complete cDNAs sequences encoding for both IGF-1 and IGF-1R in the Chilean flounder by using RT-PCR and rapid amplification of cDNAs ends (RACE) techniques. We analyzed gene expression in pre-metamorphic larvae and different organs of juvenile fish through whole mount in situ hybridization and RT-PCR, respectively. In addition, we studied the presence of calcified skeletal structures in pre-metamorphic larvae through the fluorescent chromophore calcein. The IGF-1 cDNA sequence displays an open reading frame of 558 nucleotides, encoding a 185 amino acid preproIGF-1. Moreover, IGF-1R contains an open reading frame spanning 4239 nucleotides, rendering a 702 amino acid subunit alpha and a 676 amino acid subunit beta. The deduced mature IGF-1 and IGF-1R exhibited high sequence identities with their corresponding orthologs in fishes, especially those domains involved in biological activity. RT-PCR showed expression of IGF-1 and IGF-1R transcripts in all studied tissues, consistent with their pleiotropic functions. Furthermore, we observed IGF-1 expression in notochord and IGF-1R expression in notochord, somites and head in larvae of 8 and 9 days post fertilization. Complementarily, we detected in larvae of 8 days post fertilization the presence of calcified skeletal structures in notochord and head. Interestingly, both mRNAs and calcified structures were found in territories such as notochord, an embryonic midline structure essential for the pattern of surrounding tissues as nervous system and mesoderm. Our results suggest that IGF-1 and its receptor play an important role in the development of the nervous system, muscle and bone-related structures during larval stages.


Subject(s)
Flounder/genetics , Flounder/metabolism , Gene Expression Regulation , Insulin-Like Growth Factor I , Receptor, IGF Type 1 , Animals , Chile , Cloning, Molecular , Gene Expression Profiling , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...