Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-29056479

ABSTRACT

European sea bass Dicentrarchus labrax undertake seasonal migrations to estuaries and lagoons that are characterized by fluctuations in environmental conditions. Their ability to cope with these unstable habitats is undeniable, but it is still not clear how and to what extent salinity acclimation mechanisms are affected at temperatures higher than in the sea. In this study, juvenile sea bass were pre-acclimated to seawater (SW) at 18°C (temperate) or 24°C (warm) for 2weeks and then transferred to fresh water (FW) or SW at the respective temperature. Transfer to FW for two weeks resulted in decreased blood osmolalities and plasma Cl- at both temperatures. In FW warm conditions, plasma Na+ was ~15% lower and Cl- was ~32% higher than in the temperate-water group. Branchial Na+/K+-ATPase (NKA) activity measured at the acclimation temperature (Vapparent) did not change according to the conditions. Branchial Na+/K+-ATPase activity measured at 37°C (Vmax) was lower in warm conditions and increased in FW compared to SW conditions whatever the considered temperature. Mitochondrion-rich cell (MRC) density increased in FW, notably due to the appearance of lamellar MRCs, but this increase was less pronounced in warm conditions where MRC's size was lower. In SW warm conditions, pavement cell apical microridges are less developed than in other conditions. Overall gill morphometrical parameters (filament thickness, lamellar length and width) differ between fish that have been pre-acclimated to different temperatures. This study shows that a thermal change affects gill plasticity affecting whole-organism ion balance two weeks after salinity transfer.


Subject(s)
Bass/physiology , Osmoregulation , Stress, Physiological , Animals , Aquaculture , Bass/blood , Bass/growth & development , Branchial Region/enzymology , Branchial Region/metabolism , Branchial Region/ultrastructure , Fish Proteins/metabolism , Fisheries , France , Gills/enzymology , Gills/metabolism , Gills/ultrastructure , Hot Temperature/adverse effects , Microscopy, Electron, Scanning , Salinity , Salt Tolerance , Sodium-Potassium-Exchanging ATPase/metabolism , Thermotolerance
2.
Dev Biol ; 429(1): 321-334, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28625871

ABSTRACT

Cranial neural crest cells are a multipotent cell population that generate all the elements of the pharyngeal cartilage with differentiation into chondrocytes tightly regulated by temporal intracellular and extracellular cues. Here, we demonstrate a novel role for miR-27, a highly enriched microRNA in the pharyngeal arches, as a positive regulator of chondrogenesis. Knock down of miR-27 led to nearly complete loss of pharyngeal cartilage by attenuating proliferation and blocking differentiation of pre-chondrogenic cells. Focal adhesion kinase (FAK) is a key regulator in integrin-mediated extracellular matrix (ECM) adhesion and has been proposed to function as a negative regulator of chondrogenesis. We show that FAK is downregulated in the pharyngeal arches during chondrogenesis and is a direct target of miR-27. Suppressing the accumulation of FAK in miR-27 morphants partially rescued the severe pharyngeal cartilage defects observed upon knock down of miR-27. These data support a crucial role for miR-27 in promoting chondrogenic differentiation in the pharyngeal arches through regulation of FAK.


Subject(s)
Branchial Region/embryology , Branchial Region/enzymology , Chondrogenesis/genetics , Focal Adhesion Protein-Tyrosine Kinases/metabolism , MicroRNAs/metabolism , Zebrafish/embryology , Zebrafish/genetics , Animal Fins/embryology , Animal Fins/metabolism , Animals , Cartilage/pathology , Cell Differentiation/genetics , Cell Proliferation/genetics , Cell Survival/genetics , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , MicroRNAs/genetics , Morphogenesis/genetics , Neural Crest/cytology
3.
Reprod Toxicol ; 58: 140-8, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26482727

ABSTRACT

Orofacial clefts, the most prevalent of developmental anomalies, occur with a frequency of 1 in 700 live births. Maternal cigarette smoking during pregnancy represents a risk factor for having a child with a cleft lip and/or cleft palate. Using primary cultures of first branchial arch-derived cells (1-BA cells), which contribute to the formation of the lip and palate, the present study addressed the hypothesis that components of cigarette smoke alter global DNA methylation, and/or expression of DNA methyltransferases (Dnmts) and various methyl CpG-binding proteins. Primary cultures of 1-BA cells, exposed to 80µg/mL cigarette smoke extract (CSE) for 24h, exhibited a >13% decline in global DNA methylation and triggered proteasomal-mediated degradation of Dnmts (DNMT-1 and -3a), methyl CpG binding protein 2 (MeCP2) and methyl-CpG binding domain protein 3 (MBD-3). Pretreatment of 1-BA cells with the proteasomal inhibitor MG-132 completely reversed such degradation. Collectively, these data allow the suggestion of a potential epigenetic mechanism underlying maternal cigarette smoke exposure-induced orofacial clefting.


Subject(s)
Branchial Region/enzymology , Cleft Lip/genetics , Cleft Palate/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Methyl-CpG-Binding Protein 2/metabolism , Proteasome Endopeptidase Complex/metabolism , Smoke/adverse effects , Tobacco Products/adverse effects , Transcription Factors/metabolism , Animals , Branchial Region/drug effects , Branchial Region/pathology , Cells, Cultured , Cleft Lip/enzymology , Cleft Palate/enzymology , CpG Islands , DNA (Cytosine-5-)-Methyltransferase 1 , DNA Methylation/drug effects , DNA Methyltransferase 3A , Epigenesis, Genetic/drug effects , Female , Mice, Inbred ICR , Pregnancy , Primary Cell Culture , Proteasome Inhibitors/pharmacology , Proteolysis , Risk Factors , Smoking/adverse effects
4.
Article in English | MEDLINE | ID: mdl-25193178

ABSTRACT

This study investigated the effects of two rearing salinities, and acute salinity transfer, on the energetic costs of osmoregulation and the expression of metabolic and osmoregulatory genes in the gill of Mozambique tilapia. Using automated, intermittent-flow respirometry, measured standard metabolic rates (SMRs) of tilapia reared in seawater (SW, 130 mg O2 kg⁻¹ h⁻¹) were greater than those reared in fresh water (FW, 103 mg O2 kg⁻¹ h⁻¹), when normalized to a common mass of 0.05 kg and at 25±1°C. Transfer from FW to 75% SW increased SMR within 18h, to levels similar to SW-reared fish, while transfer from SW to FW decreased SMR to levels similar to FW-reared fish. Branchial gene expression of Na⁺-K⁺-2Cl⁻ cotransporter (NKCC), an indicator of SW-type mitochondria-rich (MR) cells, was positively correlated with SMR, while Na⁺-Cl⁻ cotransporter (NCC), an indicator of FW-type MR cells, was negatively correlated. Principal Components Analysis also revealed that branchial expression of cytochrome c oxidase subunit IV (COX-IV), glycogen phosphorylase (GP), and a putative mitochondrial biogenesis regulator in fish, peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α), were correlated with a higher SMR, plasma osmolality, and environmental salinity, while expression of glycogen synthase (GS), PGC-1ß, and nuclear respiratory factor 1 (NRF-1) had negative correlations. These results suggest that the energetic costs of osmoregulation are higher in SW than in FW, which may be related to the salinity-dependent differences in osmoregulatory mechanisms found in the gills of Mozambique tilapia.


Subject(s)
Branchial Region/physiology , Energy Metabolism , Gene Expression Regulation, Developmental , Osmoregulation , Stress, Physiological , Tilapia/physiology , Animals , Aquaculture , Branchial Region/enzymology , Branchial Region/growth & development , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Fish Proteins/genetics , Fish Proteins/metabolism , Fresh Water , Gills/enzymology , Gills/growth & development , Gills/physiology , Glycogen Phosphorylase/genetics , Glycogen Phosphorylase/metabolism , Male , Principal Component Analysis , Salinity , Seawater , Solute Carrier Family 12, Member 1/genetics , Solute Carrier Family 12, Member 1/metabolism , Solute Carrier Family 12, Member 3/genetics , Solute Carrier Family 12, Member 3/metabolism , Tilapia/growth & development , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Hum Genet ; 132(5): 537-52, 2013 May.
Article in English | MEDLINE | ID: mdl-23354975

ABSTRACT

We describe a female subject (DGAP100) with a 46,X,t(X;5)(p11.3;q35.3)inv(5)(q35.3q35.1)dn, severe psychomotor retardation with hypotonia, global postnatal growth restriction, microcephaly, globally reduced cerebral volume, seizures, facial dysmorphia and cleft palate. Fluorescence in situ hybridization and whole-genome sequencing demonstrated that the X chromosome breakpoint disrupts KDM6A in the second intron. No genes were directly disrupted on chromosome 5. KDM6A is a histone 3 lysine 27 demethylase and a histone 3 lysine 4 methyltransferase. Expression of KDM6A is significantly reduced in DGAP100 lymphoblastoid cells compared to control samples. We identified nine additional cases with neurodevelopmental delay and various other features consistent with the DGAP100 phenotype with copy number variation encompassing KDM6A from microarray databases. We evaluated haploinsufficiency of kdm6a in a zebrafish model. kdm6a is expressed in the pharyngeal arches and ethmoid plate of the developing zebrafish, while a kdm6a morpholino knockdown exhibited craniofacial defects. We conclude KDM6A dosage regulation is associated with severe and diverse structural defects and developmental abnormalities.


Subject(s)
Abnormalities, Multiple/genetics , Chromosomes, Human, Pair 5 , Haploinsufficiency/genetics , Histone Demethylases/genetics , Nuclear Proteins/genetics , X Chromosome , Animals , Branchial Region/enzymology , Cell Line , Chromosomes, Human, Pair 5/genetics , Cleft Palate/genetics , Comparative Genomic Hybridization , DNA Copy Number Variations , Female , Gene Knockdown Techniques , Histone Demethylases/metabolism , Humans , Intellectual Disability/genetics , Karyotyping , Microcephaly/genetics , Muscle Hypotonia/genetics , Nuclear Proteins/metabolism , Phenotype , Psychomotor Disorders/genetics , Seizures/genetics , Translocation, Genetic , X Chromosome/genetics , Young Adult , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
6.
PLoS One ; 7(10): e47794, 2012.
Article in English | MEDLINE | ID: mdl-23094090

ABSTRACT

The neural crest is a population of multipotent cells that migrates extensively throughout vertebrate embryos to form diverse structures. Mice mutant for the de novo DNA methyltransferase DNMT3b exhibit defects in two neural crest derivatives, the craniofacial skeleton and cardiac ventricular septum, suggesting that DNMT3b activity is necessary for neural crest development. Nevertheless, the requirement for DNMT3b specifically in neural crest cells, as opposed to interacting cell types, has not been determined. Using a conditional DNMT3b allele crossed to the neural crest cre drivers Wnt1-cre and Sox10-cre, neural crest DNMT3b mutants were generated. In both neural crest-specific and fully DNMT3b-mutant embryos, cranial neural crest cells exhibited only subtle migration defects, with increased numbers of dispersed cells trailing organized streams in the head. In spite of this, the resulting cranial ganglia, craniofacial skeleton, and heart developed normally when neural crest cells lacked DNMT3b. This indicates that DNTM3b is not necessary in cranial neural crest cells for their development. We conclude that defects in neural crest derivatives in DNMT3b mutant mice reflect a requirement for DNMT3b in lineages such as the branchial arch mesendoderm or the cardiac mesoderm that interact with neural crest cells during formation of these structures.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , Gene Expression Regulation, Developmental , Neural Crest/enzymology , Neurogenesis/genetics , Animals , Basal Ganglia/embryology , Basal Ganglia/enzymology , Branchial Region/embryology , Branchial Region/enzymology , Cell Differentiation , Cell Movement , DNA (Cytosine-5-)-Methyltransferases/metabolism , Embryo, Mammalian , Heart/embryology , Integrases/genetics , Integrases/metabolism , Mesoderm/embryology , Mesoderm/enzymology , Mice , Mice, Transgenic , Mutation , Neural Crest/embryology , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , Skull/embryology , Skull/enzymology , Wnt1 Protein/genetics , Wnt1 Protein/metabolism , DNA Methyltransferase 3B
7.
Annu Rev Biochem ; 81: 429-50, 2012.
Article in English | MEDLINE | ID: mdl-22482905

ABSTRACT

[FeFe]-hydrogenses and molybdenum (Mo)-nitrogenase are evolutionarily unrelated enzymes with unique complex iron-sulfur cofactors at their active sites. The H cluster of [FeFe]-hydrogenases and the FeMo cofactor of Mo-nitrogenase require specific maturation machinery for their proper synthesis and insertion into the structural enzymes. Recent insights reveal striking similarities in the biosynthetic pathways of these complex cofactors. For both systems, simple iron-sulfur cluster precursors are modified on assembly scaffolds by the activity of radical S-adenosylmethionine (SAM) enzymes. Radical SAM enzymes are responsible for the synthesis and insertion of the unique nonprotein ligands presumed to be key structural determinants for their respective catalytic activities. Maturation culminates in the transfer of the intact cluster assemblies to a cofactor-less structural protein recipient. Required roles for nucleotide binding and hydrolysis have been implicated in both systems, but the specific role for these requirements remain unclear. In this review, we highlight the progress on [FeFe]-hydrogenase H cluster and nitrogenase FeMo-cofactor assembly in the context of these emerging paradigms.


Subject(s)
Bacteria/metabolism , Coenzymes/metabolism , Iron-Sulfur Proteins/metabolism , Iron/metabolism , Sulfur/metabolism , Bacteria/enzymology , Branchial Region/enzymology , Branchial Region/metabolism , Coenzymes/chemistry , Hydrogenase/chemistry , Hydrogenase/metabolism , Iron-Sulfur Proteins/chemistry , Molybdoferredoxin/chemistry , Molybdoferredoxin/metabolism , S-Adenosylmethionine/metabolism
8.
Mech Dev ; 128(3-4): 200-7, 2011.
Article in English | MEDLINE | ID: mdl-21256960

ABSTRACT

MicroRNAs (miRNAs) play important roles in regulating gene expression during numerous biological/pathological processes. Dicer encodes an RNase III endonuclease that is essential for generating most, if not all, functional miRNAs. In this work, we applied a conditional gene inactivation approach to examine the function of Dicer during neural crest cell (NCC) development. Mice with NCC-specific inactivation of Dicer died perinatally. Cranial and cardiac NCC migration into target tissues was not affected by Dicer disruption, but their subsequent development was disturbed. NCC derivatives and their associated mesoderm-derived cells displayed massive apoptosis, leading to severe abnormalities during craniofacial morphogenesis and organogenesis. In addition, the 4th pharyngeal arch artery (PAA) remodeling was affected, resulting in interrupted aortic arch artery type B (IAA-B) in mutant animals. Taken together, our results show that Dicer activity in NCCs is essential for craniofacial development and pharyngeal arch artery morphogenesis.


Subject(s)
Arteries/embryology , Branchial Region/embryology , Craniofacial Abnormalities/embryology , DEAD-box RNA Helicases/metabolism , Neural Crest/cytology , Ribonuclease III/metabolism , Animals , Apoptosis , Arteries/enzymology , Bone and Bones/embryology , Bone and Bones/enzymology , Branchial Region/blood supply , Branchial Region/enzymology , Cartilage/embryology , Cartilage/enzymology , Cell Movement , Cell Proliferation , Craniofacial Abnormalities/enzymology , Craniofacial Abnormalities/genetics , DEAD-box RNA Helicases/genetics , Female , Gene Silencing , Head/embryology , Head/innervation , Male , Mice , Mice, Transgenic , Morphogenesis , Muscle, Skeletal/embryology , Muscle, Skeletal/enzymology , Neural Crest/enzymology , Organogenesis , Peripheral Nervous System/embryology , Peripheral Nervous System/enzymology , Retrognathia/embryology , Retrognathia/enzymology , Retrognathia/genetics , Ribonuclease III/genetics
9.
Article in English | MEDLINE | ID: mdl-19699315

ABSTRACT

The weatherloach, Misgurnus anguillicaudatus, is a freshwater, facultative air-breathing fish that lives in streams and rice paddy fields, where it may experience drought and/or high environmental ammonia (HEA) conditions. The aim of this study was to determine what roles branchial Na(+)/K(+)-ATPase, H(+)-ATPase, and Rhcg have in ammonia tolerance and how the weatherloach copes with ammonia loading conditions. The loach's high ammonia tolerance was confirmed as was evident from its high 96 h LC(50) value and high tissue tolerance to ammonia. The weatherloach does not appear to make use of Na(+)/NH(4)(+)-ATPase facilitated transport to excrete ammonia when exposed to HEA or to high environmental pH since no changes in activity were observed. Using immunofluorescence microscopy, distinct populations of vacuolar (V)-type H(+)-ATPase and Na(+)/K(+)-ATPase immunoreactive cells were identified in branchial epithelia, with apical and basolateral staining patterns, respectively. Rhesus C glycoprotein (Rhcg1), an ammonia transport protein, immunoreactivity was also found in a similar pattern as H(+)-ATPase. Rhcg1 (Slc42a3) mRNA expression also increased significantly during aerial exposure, although not significantly under ammonia loading conditions. The colocalization of H(+)-ATPase and Rhcg1 to the similar non-Na(+)/K(+)-ATPase immunoreactive cell type would support a role for H(+)-ATPase in ammonia excretion via Rhcg by NH(4)(+) trapping. The importance of gill boundary layer acidification in net ammonia excretion was confirmed in this fish; however, it was not associated with an increase in H(+)-ATPase expression, since tissue activity and protein levels did not increase with high environmental pH and/or HEA. However the V-ATPase inhibitor, bafilomycin, did decrease net ammonia flux whereas other ion transport inhibitors (amiloride, SITS) had no effect. H(+)-ATPase inhibition also resulted in a consequent elevation in plasma ammonia levels and a decrease in the net acid flux. In gill, aerial exposure was also associated with a significant increase in membrane fluidity (or increase in permeability) which would presumably enhance NH(3) permeation through the plasma membrane. Taken together, these results indicate the gill of the weatherloach is responsive to aerial conditions that would aid ammonia excretion.


Subject(s)
Ammonia/metabolism , Branchial Region/metabolism , Cypriniformes/metabolism , Gills/metabolism , Adaptation, Physiological , Animals , Biological Transport , Branchial Region/enzymology , Cation Transport Proteins/metabolism , Cell Membrane Permeability , Cypriniformes/genetics , Fish Proteins/metabolism , Fluorescent Antibody Technique , Gills/enzymology , Hydrogen-Ion Concentration , Kinetics , Membrane Fluidity , Muscles/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sodium-Potassium-Exchanging ATPase/metabolism , Vacuolar Proton-Translocating ATPases/metabolism
10.
Toxicol Sci ; 113(1): 70-6, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19833739

ABSTRACT

Biotransformation of inorganic arsenic (iAs) involves methylation catalyzed by arsenic (+ 3 oxidation state) methyltransferase (As3mt) yielding mono-, di-, and trimethylated arsenicals. To investigate the evolution of molecular mechanisms that mediate arsenic biotransformation, a comparative genomic approach focusing on the invertebrate chordate Ciona intestinalis was used. Bioinformatic analyses identified an As3mt gene in the C. intestinalis genome. Constitutive As3mt RNA expression was observed in heart, branchial sac, and gastrointestinal tract. Adult animals were exposed to 0 or 1 ppm of iAs for 1 or 5 days. Steady-state As3mt RNA expression in the gastrointestinal tract was not modulated significantly by 5 days of exposure to iAs. Tissue levels of iAs and its methylated metabolites were determined by hydride generation-cryotrapping-gas chromatography-atomic absorption spectrometry. At either time point, exposure to iAs significantly increased concentrations of iAs and its methylated metabolites in tissues. After 5 days of exposure, total speciated arsenic concentrations were highest in branchial sac (3705 ng/g), followed by heart (1019 ng/g) and gastrointestinal tract (835 ng/g). At this time point, the sum of the speciated arsenical concentrations in gastrointestinal tract and heart equaled or exceeded that of iAs; in branchial sac, iAs was the predominant species present. Ciona intestinalis metabolizes iAs to its methylated metabolites, which are retained in tissues. This metabolic pattern is consistent with the presence of an As3mt ortholog in its genome and constitutive expression of the gene in prominent organs, making this basal chordate a useful model to examine the evolution of arsenic detoxification.


Subject(s)
Arsenites/metabolism , Ciona intestinalis/enzymology , Methyltransferases/metabolism , Sodium Compounds/metabolism , Water Pollutants, Chemical/metabolism , Amino Acid Sequence , Animals , Arsenites/toxicity , Biotransformation , Branchial Region/enzymology , Chromatography, Gas , Ciona intestinalis/drug effects , Ciona intestinalis/genetics , Databases, Genetic , Evolution, Molecular , Gastrointestinal Tract/enzymology , Gene Expression Regulation, Enzymologic , Methylation , Methyltransferases/genetics , Molecular Sequence Data , Myocardium/enzymology , Oxidation-Reduction , RNA, Messenger/metabolism , Sodium Compounds/toxicity , Spectrophotometry, Atomic , Time Factors , Water Pollutants, Chemical/toxicity
11.
Development ; 135(22): 3775-87, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18927157

ABSTRACT

Skeletal syndromes are among the most common birth defects. Vertebrate skeletogenesis involves two major cell types: cartilage-forming chondrocytes and bone-forming osteoblasts. In vitro, both are under the control of retinoic acid (RA), but its exact in vivo effects remained elusive. Here, based on the positional cloning of the dolphin mutation, we have studied the role of the RA-oxidizing enzyme Cyp26b1 during cartilage and bone development in zebrafish. cyp26b1 is expressed in condensing chondrocytes as well as in osteoblasts and their precursors. cyp26b1 mutants and RA-treated wild-type fish display a reduction in midline cartilage and the hyperossification of facial and axial bones, leading to fusions of vertebral primordia, a defect not previously described in the context of RA signaling. Fusions of cervical vertebrae were also obtained by treating mouse fetuses with the specific Cyp26 inhibitor R115866. Together with data on the expression of osteoblast markers, our results indicate that temporal and spatial restriction of RA signaling by Cyp26 enzymes is required to attenuate osteoblast maturation and/or activity in vivo. cyp26b1 mutants may serve as a model to study the etiology of human vertebral disorders such as Klippel-Feil anomaly.


Subject(s)
Body Patterning , Cytochrome P-450 Enzyme System/metabolism , Gene Expression Regulation, Developmental , Osteogenesis , Tretinoin/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Base Sequence , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Branchial Region/embryology , Branchial Region/enzymology , Cytochrome P-450 Enzyme System/genetics , Gene Expression Regulation, Enzymologic , Mice , Mutation/genetics , Osteoblasts/enzymology , Osteopontin/metabolism , Phenotype , Retinoic Acid 4-Hydroxylase , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
12.
Article in English | MEDLINE | ID: mdl-18571989

ABSTRACT

California Mozambique tilapia (Oreochromis mossambicus x O. urolepis hornorum) are extremely saline tolerant and have been previously shown to reduce whole-animal oxygen consumption rate (MO(2)) upon exposures to salinities greater than that of seawater (SW). In this study tilapia were acclimated to 15, 30, 45, 60 and 75 g/L salinity for 1, 5, 14, or 28 days. There was little change in plasma osmolality or muscle water content in salinities below 60 g/L, and branchial Na(+), K(+)-ATPase (NKA) activity was low in 15 and 30 g/L relative to 60 and 75 g/L. When tilapia were exposed to 75 g/L, plasma osmolality and NKA activity were significantly increased within 5 days of exposure relative to those in 15 and 30 g/L, and remained elevated over the entire 28 days acclimation, indicating that short term salinity challenges (i.e., 5 days) are predictive of longer exposure durations in this species. MO(2) following transfer to 15 and 30 g/L was elevated, reflecting the high energy demand required for switching from a hyper- to a hypo-osmoregulatory strategy. The MO(2) of 60 g/L-exposed fish was significantly reduced at 1, 5, and 14 days, relative to 30 g/L-exposed fish; however by 28 days there were no significant differences. We investigated the potential for a metabolic basis for the salinity-induced MO(2) reduction, using forward stepwise linear regression to correlate enzyme activities of brain, liver, and kidney with MO(2). Brain NKA was correlated with MO(2) after 5 days (p<0.01, r(2)=0.944) and both brain NKA and hepatic total ATPase were correlated with the reduced MO(2) at 14 days (p=0.027, r(2)=0.980 and p=0.025, r(2)=0.780, respectively). These results may indicate a tissue-level metabolic suppression, which has not been previously described as a response to hypersaline exposure in fishes.


Subject(s)
Energy Metabolism , Fresh Water/chemistry , Salinity , Seawater/chemistry , Tilapia/metabolism , Water-Electrolyte Balance , Acclimatization , Animals , Brain/enzymology , Branchial Region/enzymology , Down-Regulation , Intestines/enzymology , Kidney/enzymology , Kinetics , Liver/enzymology , Muscles/metabolism , Osmolar Concentration , Oxygen Consumption , Sodium-Potassium-Exchanging ATPase/metabolism , Tilapia/blood
13.
Article in English | MEDLINE | ID: mdl-17347004

ABSTRACT

Chloride transport mechanisms in the gills of the estuarine spotted green pufferfish (Tetraodon nigroviridis) were investigated. Protein abundance of Na(+)/K(+)-ATPase (NKA) and the other four chloride transporters, i.e., Na(+)/K(+)/2Cl(-) cotransporter (NKCC), cystic fibrosis transmembrane conductance regulator (CFTR), Cl(-)/HCO(3)(-) anion exchanger 1 (AE1), and chloride channel 3 (CLC-3) in gills of the seawater- (SW; 35 per thousand) or freshwater (FW)-acclimatized fish were examined by immunoblot analysis. Appropriate negative controls were used to confirm the specificity of the antibodies to the target proteins. The relative protein abundance of NKA was higher (i.e., 2-fold) in gills of the SW group compared to the FW group. NKCC and CFTR were expressed in gills of the SW group but not in the FW group. In contrast, the levels of relative protein abundance of branchial AE1 and CLC-3 in the FW group were 23-fold and 2.7-fold higher, respectively, compared to those of the SW group. This study is first of its kind to provide direct in vivo evidence of the protein expression of CLC-3 in teleostean gills, as well as to examine the simultaneous protein expression of the Cl(-) transporters, especially AE1 and CLC-3 of FW- and SW-acclimatized teleosts. The differential protein expression of NKA, chloride transporters in gills of the FW- and SW-acclimatized T. nigroviridis observed in the present study shows their close relationship to the physiological homeostasis (stable blood osmolality), as well as explains the impressive ionoregulatory ability of this euryhaline species in response to salinity challenges.


Subject(s)
Anion Exchange Protein 1, Erythrocyte/metabolism , Chloride Channels/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Gills/enzymology , Sodium-Potassium-Chloride Symporters/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Tetraodontiformes/metabolism , Animals , Branchial Region/drug effects , Branchial Region/enzymology , Environment , Erythrocytes/drug effects , Erythrocytes/metabolism , Gills/drug effects , Sodium Chloride/pharmacology
14.
Article in English | MEDLINE | ID: mdl-17350299

ABSTRACT

The Pacific white shrimp, Litopenaeus vannamei, acclimated to 30 ppt salinity, was transferred to either low (15 and 5 ppt), or high (45 ppt) salinity for 7 days. Hemolymph osmolality, branchial carbonic anhydrase activity, and total ninhydrin-positive substances (TNPS) in abdominal muscle were then measured for each condition. Hemolymph osmotic concentration was regulated slightly below ambient water osmolality in shrimp acclimated to 30 ppt. At 15 and 5 ppt, shrimp were strong hyper-osmotic regulators, maintaining hemolymph osmolality between 200 and 400 mOsm above ambient. Shrimp acclimated to 30 ppt and transferred to 45 ppt salinity were strong hypo-osmotic and hypo-ionic regulators, maintaining hemolymph osmolality over 400 mOsm below ambient. Branchial carbonic anhydrase (CA) activity was low (approximately 100 micromol CO(2) mg protein(-1) min(-1)) and uniform across all 8 gills in shrimp acclimated to 30 ppt, but CA activity increased in all gills after exposure to both low and high salinities. Anterior gills had the largest increases in CA activity, and levels of increase were approximately the same for low and high salinity exposure. Branchial CA induction appears to be functionally important in both hyper- and hypo-osmotic regulations of hemolymph osmotic concentrations. Abdominal muscle TNPS made up between 19 and 38% of the total intracellular osmotic concentration in shrimp acclimated to 5, 15, and 30 ppt. TNPS levels did not change across this salinity range, over which hemolymph osmotic concentrations were tightly regulated. At 45 ppt, hemolymph osmolality increased, and muscle TNPS also increased, presumably to counteract intracellular water loss and restore cell volume. L. vannamei appears to employ mechanisms of both extracellular osmoregulation and intracellular volume regulation as the basis of its euryhalinity.


Subject(s)
Acclimatization/drug effects , Branchial Region/enzymology , Carbonic Anhydrases/metabolism , Decapoda/drug effects , Decapoda/enzymology , Ninhydrin/metabolism , Sodium Chloride/pharmacology , Animals , Branchial Region/drug effects , Decapoda/physiology , Gills/drug effects , Gills/metabolism , Hemolymph/chemistry , Hemolymph/drug effects , Muscles/chemistry , Muscles/drug effects , Ninhydrin/analysis , Osmolar Concentration , Pacific Ocean
15.
Dev Biol ; 304(1): 194-207, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17239364

ABSTRACT

Genetic and pharmacological studies demonstrate that Endothelin1 (Edn1) is a key signaling molecule for patterning the facial skeleton in fish, chicks, and mice. When Edn1 function is reduced early in development the ventral lower jaw and supporting structures are reduced in size and often fused to their dorsal upper jaw counterparts. We show that schmerle (she) encodes a zebrafish ortholog of Phospholipase C, beta 3 (Plcbeta3) required in cranial neural crest cells for Edn1 regulation of pharyngeal arch patterning. Sequencing and co-segregation demonstrates that two independent she (plcbeta3) alleles have missense mutations in conserved residues within the catalytic domains of Plcbeta3. Homozygous plcbeta3 mutants are phenotypically similar to edn1 mutants and exhibit a strong arch expression defect in Edn1-dependent Distalless (Dlx) genes as well as expression defects in several Edn1-dependent intermediate and ventral arch domain transcription factors. plcbeta3 also genetically interacts with edn1, supporting a model in which Edn1 signals through a G protein-coupled receptor to activate Plcbeta3. Mild skeletal defects occur in plcbeta3 heterozygotes, showing the plcbeta3 mutations are partially dominant. Through a morpholino-mediated deletion in the N-terminal PH domain of Plcbeta3, we observe a partial rescue of facial skeletal defects in homozygous plcbeta3 mutants, supporting a hypothesis that an intact PH domain is necessary for the partial dominance we observe. In addition, through mosaic analyses, we show that wild-type neural crest cells can efficiently rescue facial skeletal defects in homozygous plcbeta3 mutants, demonstrating that Plcbeta3 function is required in neural crest cells and not other cell types to pattern the facial skeleton.


Subject(s)
Body Patterning/physiology , Branchial Region/embryology , Endothelin-1/metabolism , Gene Expression Regulation, Developmental , Isoenzymes/genetics , Neural Crest/metabolism , Type C Phospholipases/genetics , Zebrafish Proteins/genetics , Zebrafish/embryology , Animals , Base Sequence , Branchial Region/enzymology , DNA Primers , Isoenzymes/metabolism , Microscopy, Confocal , Molecular Sequence Data , Mutation, Missense/genetics , Neural Crest/embryology , Phospholipase C beta , Sequence Analysis, DNA , Signal Transduction/genetics , Type C Phospholipases/metabolism , Zebrafish Proteins/metabolism
16.
Article in English | MEDLINE | ID: mdl-15649768

ABSTRACT

The effects of ovine prolactin (oPRL) and striped bass prolactin (sbPRL; Morone saxatilis) on plasma osmolality, electrolyte balance, and gill Na(+),K(+)-ATPase activity were investigated in hypophysectomized (Hx), freshwater (FW)-acclimated, hybrid striped bass (M. saxatilisxMorone chrysops). They were kept in dilute (isoosmotic) seawater for about 10 days after surgery. Seven days after transfer to FW, Hx fish had lower plasma osmolality and lower levels of Na(+), Cl(-), and Ca(2+) than sham-operated and intact fish. Fish were injected four times with oPRL (1, 5, or 20 microg/g body mass), sbPRL (10 or 100 ng/g), or hormone vehicle (0.9% NaCl) at 48-h intervals (days 0, 2, 4, and 6) in FW and then sampled for blood plasma 24 h after the fourth injection (day 7). In Hx fish, oPRL (5 and 20 microg/g) and sbPRL (10 and 100 ng/g) were effective in maintaining plasma osmolality and levels of Na(+), Cl(-), and Ca(2+) above values seen in saline-injected controls. Hypophysectomy did not affect branchial Na(+),K(+)-ATPase activity, but enzyme activity was significantly reduced in Hx fish receiving oPRL (20 mug/g) or sbPRL (10 or 100 ng/g). These results indicate that PRL acts to maintain plasma osmotic and ionic balance in FW-adapted hybrid striped bass, and that this may involve downregulation of branchial Na(+),K(+)-ATPase activity.


Subject(s)
Bass/physiology , Hypophysectomy , Prolactin/pharmacology , Adenosine Triphosphatases/metabolism , Animals , Bass/blood , Bass/metabolism , Branchial Region/drug effects , Branchial Region/enzymology , Electrolytes/blood , Injections , Osmolar Concentration , Prolactin/administration & dosage , Water-Electrolyte Balance/drug effects
17.
Am J Physiol Cell Physiol ; 284(1): C233-41, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12388091

ABSTRACT

Although euryhaline teleosts can adapt to environmental fluctuation of salinity, their energy source for responding to changes in salinity and osmolarity remains unclear. This study examines the cellular localization of creatine kinase (CK) expression in branchia of tilapia (Oreochromis mossambicus). Western blot analysis of muscle-type CK (MM form) revealed a high association with salinity changes, but BB and MB forms of CK in the gills of fish adapted to seawater did not change. With the use of immunocytochemistry, three CK isoforms (MM, MB, and BB) were localized in mitochondria-rich (MR) cells and other epithelial cells of tilapia gills. In addition, staining intensity of MM-form CK in MR cells increased after seawater transfer, whereas BB and MB forms did not significantly change. To our knowledge, this work presents the first evidence of CK expression in MR cells of tilapia gills, highlighting the potential role of CK in providing energy for ion transport.


Subject(s)
Branchial Region/cytology , Branchial Region/enzymology , Creatine Kinase/biosynthesis , Tilapia , Animals , Isoenzymes/biosynthesis
18.
Article in English | MEDLINE | ID: mdl-12020661

ABSTRACT

Information about the presence and effects of nitric oxide (NO) in fish vasculature is scant and contradictory. We have studied the NO/cGMP system in the branchial circulation of the teleost Anguilla anguilla using a branchial basket preparation under basal conditions and cholinergic stimulation. The effects of endogenous and exogenous NO were tested with L-arginine, the nitric oxide synthase (NOS) substrate, and the NO donors 3-morpholinosydnonimine (SIN-1) and sodium nitroprusside (SNP), respectively. L-arginine (from 10(-11) to 10(-6) M) and the NO donors (starting from 10(-14) M) caused dose-dependent vasoconstriction. Conversely, in the ACh-pre-contracted preparations both donors elicited vasodilation. SIN-1-induced vasoconstriction was due to NO generation: it was increased by superoxide dismutase (SOD) and blocked by NO scavenger hemoglobin. Pre-treatment with sGC inhibitor 1H-[1,2,4]oxadiazolo-[4,3-a]quinoxalin-1-one (ODQ) inhibited the effects of SIN-1 and SNP. The stable cGMP analogue 8-bromo-guanosine 3',5'-cyclic monophosphate (8-Br cGMP) induced dose-dependent vasoconstriction. Unexpectedly, three NOS inhibitors, N(G)-nitro-L-arginine methyl ester (L-NAME), N(G)-monomethyl-L-arginine (L-NMMA), L-N(5)-(1-iminoethyl) ornithine (L-NIO), caused mild vasoconstriction. ACh caused vasoconstriction, but at pico- and nanomolar concentrations it caused mild but significant vasodilation in 40% of the preparations. Both responses, blocked by atropine and pirenzepine, required an intact endothelium. The ACh-induced vasoconstriction was substantially independent of a NO-cGMP mechanism.


Subject(s)
Acetylcholine/pharmacology , Branchial Region/blood supply , Branchial Region/drug effects , Cyclic GMP/metabolism , Eels/physiology , Molsidomine/analogs & derivatives , Nitric Oxide/metabolism , Vasoconstriction/drug effects , Animals , Arginine/pharmacology , Branchial Region/enzymology , Branchial Region/metabolism , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Female , Gills/blood supply , Gills/drug effects , Gills/enzymology , Gills/metabolism , Hemoglobins/pharmacology , In Vitro Techniques , Male , Molsidomine/pharmacology , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism
19.
Comp Biochem Physiol B Biochem Mol Biol ; 130(3): 323-30, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11567894

ABSTRACT

The rapid action of cortisol and corticosterone on branchial Na(+)-K(+) ATPase, Ca(2+) ATPase activity and Na(+), K(+) and Ca(2+) ion contents was studied both in vivo and in vitro employing transcription inhibitor actinomycin D in Oreochromis mossambicus. Cortisol and corticosterone administration had significantly increased the activity of branchial Na(+)-K(+) ATPase and Ca(2+) ATPase in vivo after 30 min of injection, and the trend continued for 60 and 120 min for cortisol. The ionic contents were also significantly increased after 30 min in vivo. Na(+)-K(+) ATPase activity was significantly increased 5 min after hormone application in the in vitro system. Actinomycin D did not inhibit the effect of glucocorticoids on ATPase activity both in vivo and in vitro. It is concluded from the present study that cortisol and corticosterone produced a rapid stimulatory effect on branchial ATPase activity and ions in O. mossambicus both in vivo and in vitro. This effect could be due to a non-genomic action of these hormones since the enzyme activity was insensitive to actinomycin D.


Subject(s)
Adenosine Triphosphatases/metabolism , Branchial Region/drug effects , Branchial Region/enzymology , Corticosterone/pharmacology , Hydrocortisone/pharmacology , Tilapia/metabolism , Animals , Calcium/metabolism , Calcium-Transporting ATPases/metabolism , Dactinomycin/pharmacology , In Vitro Techniques , Potassium/metabolism , Sodium/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Time Factors
20.
Biochem Biophys Res Commun ; 285(5): 1338-43, 2001 Aug 03.
Article in English | MEDLINE | ID: mdl-11478804

ABSTRACT

We have isolated a novel acetyltransferase from Xenopus laevis, named Xat-1. Xat-1 cDNA encodes a predicted protein of 846 amino acids that contains tetratricopeptide repeat (TPR) domains mediating protein-protein interactions and a bipartite nuclear localization signal (NLS). Its apparent molecular mass of 98.8 kDa was determined by SDS-PAGE analysis of Xat-1 recombinant protein in vitro translated in rabbit reticulocyte lysate. Xat-1 is homologous to N-terminal acetyltransferase 1 (NAT1), a gene that was originally discovered in yeast. Furthermore, it has many orthologs from human, mouse, Drosophila, C. elegans, and even Arabidopsis, thereby suggesting that these constitute a novel acetyltransferase family whose functions have been not examined. Xat-1 transcripts are expressed at relatively constant levels throughout early embryonic stages. They also exhibit dynamic expression pattern in brain, somites, branchial arches, pronephros, and otic vesicles.


Subject(s)
Acetyltransferases/genetics , Gene Expression Regulation, Developmental , Xenopus Proteins , Acetyltransferases/chemistry , Acetyltransferases/metabolism , Animals , Brain/embryology , Brain/enzymology , Branchial Region/embryology , Branchial Region/enzymology , Cell-Free System , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Ear, Inner/embryology , Ear, Inner/enzymology , Electrophoresis, Polyacrylamide Gel , In Situ Hybridization , Kidney/embryology , Kidney/enzymology , Molecular Sequence Data , Molecular Weight , Multigene Family , Organ Specificity/genetics , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Somites/enzymology , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...