Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Physiol Genomics ; 51(1): 27-41, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30540547

ABSTRACT

Although cystic fibrosis (CF) is attributed to dysfunction of a single gene, the relationships between the abnormal gene product and the development of inflammation and progression of lung disease are not fully understood, which limits our ability to predict an individual patient's clinical course and treatment response. To better understand CF progression, we characterized the molecular signatures of CF disease status with plasma-based functional genomics. Peripheral blood mononuclear cells (PBMCs) from healthy donors were cultured with plasma samples from CF patients ( n = 103) and unrelated, healthy controls ( n = 31). Gene expression levels were measured with an Affymetrix microarray (GeneChip Human Genome U133 Plus 2.0). Peripheral blood samples from a subset of the CF patients ( n = 40) were immunophenotyped by flow cytometry, and the data were compared with historical data for age-matched healthy controls ( n = 351). Plasma samples from another subset of CF patients ( n = 56) and healthy controls ( n = 16) were analyzed by multiplex enzyme-linked immunosorbent assay (ELISA) for numerous cytokines and chemokines. Principal component analysis and hierarchical clustering of induced transcriptional data revealed disease-specific plasma-induced PBMC profiles. Among 1,094 differentially expressed probe sets, 51 genes were associated with pancreatic sufficient status, and 224 genes were associated with infection with Pseudomonas aeruginosa. The flow cytometry and ELISA data confirmed that various immune modulators are relevant contributors to the CF molecular signature. This study provides strong evidence for distinct molecular signatures among CF patients. An understanding of these molecular signatures may lead to unique molecular markers that will enable more personalized prognoses, individualized treatment plans, and rapid monitoring of treatment response.


Subject(s)
Cystic Fibrosis/blood , Cystic Fibrosis/genetics , Plasma/metabolism , Transcriptome/genetics , Adolescent , Adult , Blood Donors , Case-Control Studies , Child , Child, Preschool , Cohort Studies , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cytokines/blood , Female , Genotype , Humans , Immunophenotyping , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Mutation , Neutrophils/metabolism , Oligonucleotide Array Sequence Analysis , Reactive Oxygen Species/metabolism , Young Adult
2.
PLoS One ; 10(7): e0132474, 2015.
Article in English | MEDLINE | ID: mdl-26192751

ABSTRACT

In the heart, scaffolding proteins such as A-Kinase Anchoring Proteins (AKAPs) play a crucial role in normal cellular function by serving as a signaling hub for multiple protein kinases including protein kinase D1 (PKD1). Under cardiac hypertrophic conditions AKAP13 anchored PKD1 activates the transcription factor MEF2 leading to subsequent fetal gene activation and hypertrophic response. We used an expression microarray to identify the global transcriptional response in the hearts of wild-type mice expressing the native form of AKAP13 compared to a gene-trap mouse model expressing a truncated form of AKAP13 that is unable to bind PKD1 (AKAP13-ΔPKD1). Microarray analysis showed that AKAP13-ΔPKD1 mice broadly failed to exhibit the transcriptional profile normally associated with compensatory cardiac hypertrophy following trans-aortic constriction (TAC). The identified differentially expressed genes in WT and AKAP13-ΔPKD1 hearts are vital for the compensatory hypertrophic response to pressure-overload and include myofilament, apoptotic, and cell growth/differentiation genes in addition to genes not previously identified as affected by AKAP13-anchored PKD1. Our results show that AKAP13-PKD1 signaling is critical for transcriptional regulation of key contractile, cell death, and metabolic pathways during the development of compensatory hypertrophy in vivo.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cardiomegaly/metabolism , Gene Expression Regulation , Guanine Nucleotide Exchange Factors/metabolism , Protein Kinase C/metabolism , Signal Transduction/genetics , A Kinase Anchor Proteins/genetics , Animals , Cardiomegaly/genetics , Cell Death/genetics , Gene Expression Profiling , Guanine Nucleotide Exchange Factors/genetics , Mice , Mice, Transgenic , Minor Histocompatibility Antigens , Myocardial Contraction/genetics , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Protein Kinase C/genetics , Transcription, Genetic
3.
Cell Signal ; 27(5): 908-22, 2015 May.
Article in English | MEDLINE | ID: mdl-25683917

ABSTRACT

Hypertrophy increases the risk of heart failure and arrhythmia. Prevention or reversal of the maladaptive hypertrophic phenotype has thus been proposed to treat heart failure. Chronic ß-adrenergic receptor (ß-AR) stimulation induces cardiomyocyte hypertrophy by elevating 3',5'-cyclic adenosine monophosphate (cAMP) levels and activating downstream effectors such protein kinase A (PKA). Conversely, hydrolysis of cAMP by phosphodiesterases (PDEs) spatiotemporally restricts cAMP signaling. Here, we demonstrate that PDE4, but not PDE3, is critical in regulating cardiomyocyte hypertrophy, and may represent a potential target for preventing maladaptive hypertrophy. We identify a sequence within the upstream conserved region 1 of PDE4D, termed UCR1C, as a novel activator of PDE4 long isoforms. UCR1C activates PDE4 in complex with A-kinase anchoring protein (AKAP)-Lbc resulting in decreased PKA signaling facilitated by AKAP-Lbc. Expression of UCR1C in cardiomyocytes inhibits hypertrophy in response to chronic ß-AR stimulation. This effect is partially due to inhibition of nuclear PKA activity, which decreases phosphorylation of the transcription factor cAMP response element-binding protein (CREB). In conclusion, PDE4 activation by UCR1C attenuates cardiomyocyte hypertrophy by specifically inhibiting nuclear PKA activity.


Subject(s)
Cardiomegaly/drug therapy , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Enzyme Activation/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Peptides/chemistry , Peptides/pharmacology , A Kinase Anchor Proteins/metabolism , Amino Acid Sequence , Animals , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , HEK293 Cells , Humans , Molecular Sequence Data , Myocytes, Cardiac/metabolism , Phosphorylation/drug effects , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Rats , Signal Transduction/drug effects
4.
PLoS One ; 9(1): e85000, 2014.
Article in English | MEDLINE | ID: mdl-24465463

ABSTRACT

Serine-threonine Protein phosphatase 2 A (PP2A), a member of the PPP family of phosphatases, regulates a variety of essential cellular processes, including cell-cycling, DNA replication, transcription, translation, and secondary signaling pathways. In the heart, increased PP2A activity/signaling has been linked to cardiac remodeling, contractile dysfunction and, in failure, arrythmogenicity. The core PP2A complex is a hetero-trimeric holoenzyme consisting of a 36 kDa catalytic subunit (PP2Ac); a regulatory scaffold subunit of 65 kDa (PR65A or PP2Aa); and one of at least 18 associated variable regulatory proteins (B subunits) classified into 3 families. In the present study, three in vivo sites of phosphorylation in cardiac PR65A are identified (S303, T268, S314). Using HEK cells transfected with recombinant forms of PR65A with phosphomimetic (P-PR65A) and non-phosphorylated (N-PR65A) amino acid substitutions at these sites, these phosphorylations were shown to inhibit the interaction of PR65A with PP2Ac and PP2A holoenzyme signaling. Forty-seven phospho-proteins were increased in abundance in HEK cells transfected with P-PR65A versus N-PR65A by phospho-protein profiling using 2D-DIGE analysis on phospho-enriched whole cell protein extracts. Among these proteins were elongation factor 1α (EF1A), elongation factor 2, heat shock protein 60 (HSP60), NADPH-dehydrogenase 1 alpha sub complex, annexin A, and PR65A. Compared to controls, failing hearts from the Dahl rat had less phosphorylated PR65A protein abundance and increased PP2A activity. Thus, PR65A phosphorylation is an in vivo mechanism for regulation of the PP2A signaling complex and increased PP2A activity in heart failure.


Subject(s)
Heart Failure/metabolism , Myocardium/metabolism , Protein Phosphatase 2/metabolism , Protein Subunits/metabolism , Signal Transduction , Animals , Annexin A1/genetics , Annexin A1/metabolism , Chaperonin 60/genetics , Chaperonin 60/metabolism , Gene Expression Regulation , HEK293 Cells , Heart Failure/genetics , Heart Failure/pathology , Humans , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Myocardium/pathology , NADPH Dehydrogenase/genetics , NADPH Dehydrogenase/metabolism , Peptide Elongation Factor 1/genetics , Peptide Elongation Factor 1/metabolism , Peptide Elongation Factor 2/genetics , Peptide Elongation Factor 2/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Protein Multimerization , Protein Phosphatase 2/genetics , Protein Subunits/genetics , Rats , Rats, Inbred Dahl
5.
J Mol Cell Cardiol ; 66: 27-40, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24161911

ABSTRACT

The objective of this study was to determine the role of A-Kinase Anchoring Protein (AKAP)-Lbc in the development of heart failure, by investigating AKAP-Lbc-protein kinase D1 (PKD1) signaling in vivo in cardiac hypertrophy. Using a gene-trap mouse expressing a truncated version of AKAP-Lbc (due to disruption of the endogenous AKAP-Lbc gene), that abolishes PKD1 interaction with AKAP-Lbc (AKAP-Lbc-ΔPKD), we studied two mouse models of pathological hypertrophy: i) angiotensin (AT-II) and phenylephrine (PE) infusion and ii) transverse aortic constriction (TAC)-induced pressure overload. Our results indicate that AKAP-Lbc-ΔPKD mice exhibit an accelerated progression to cardiac dysfunction in response to AT-II/PE treatment and TAC. AKAP-Lbc-ΔPKD mice display attenuated compensatory cardiac hypertrophy, increased collagen deposition and apoptosis, compared to wild-type (WT) control littermates. Mechanistically, reduced levels of PKD1 activation are observed in AKAP-Lbc-ΔPKD mice compared to WT mice, resulting in diminished phosphorylation of histone deacetylase 5 (HDAC5) and decreased hypertrophic gene expression. This is consistent with a reduced compensatory hypertrophy phenotype leading to progression of heart failure in AKAP-Lbc-ΔPKD mice. Overall, our data demonstrates a critical in vivo role for AKAP-Lbc-PKD1 signaling in the development of compensatory hypertrophy to enhance cardiac performance in response to TAC-induced pressure overload and neurohumoral stimulation by AT-II/PE treatment.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cardiomegaly/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Heart Failure/metabolism , Myocardium/metabolism , Protein Kinase C/metabolism , A Kinase Anchor Proteins/chemistry , A Kinase Anchor Proteins/genetics , Angiotensin II/adverse effects , Animals , Aorta/pathology , Apoptosis , Cardiomegaly/chemically induced , Cardiomegaly/genetics , Cardiomegaly/pathology , Collagen/genetics , Collagen/metabolism , Female , Gene Expression Regulation , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/genetics , Heart Failure/chemically induced , Heart Failure/genetics , Heart Failure/pathology , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Male , Mice , Mice, Transgenic , Minor Histocompatibility Antigens , Myocardium/pathology , Phenylephrine/adverse effects , Protein Kinase C/genetics , Protein Structure, Tertiary , Signal Transduction
6.
BMC Evol Biol ; 12: 125, 2012 Jul 26.
Article in English | MEDLINE | ID: mdl-22834419

ABSTRACT

BACKGROUND: A-Kinase Anchoring Proteins (AKAPs) are molecular scaffolding proteins mediating the assembly of multi-protein complexes containing cAMP-dependent protein kinase A (PKA), directing the kinase in discrete subcellular locations. Splice variants from the AKAP7 gene (AKAP15/18) are vital components of neuronal and cardiac phosphatase complexes, ion channels, cardiac Ca2+ handling and renal water transport. RESULTS: Shown in evolutionary analyses, the formation of the AKAP7-RI/RII binding domain (required for AKAP/PKA-R interaction) corresponds to vertebrate-specific gene duplication events in the PKA-RI/RII subunits. Species analyses of AKAP7 splice variants shows the ancestral AKAP7 splice variant is AKAP7α, while the ancestral long form AKAP7 splice variant is AKAP7γ. Multi-species AKAP7 gene alignments, show the recent formation of AKAP7δ occurs with the loss of native AKAP7γ in rats and basal primates. AKAP7 gene alignments and two dimensional Western analyses indicate that AKAP7γ is produced from an internal translation-start site that is present in the AKAP7δ cDNA of mice and humans but absent in rats. Immunofluorescence analysis of AKAP7 protein localization in both rat and mouse heart suggests AKAP7γ replaces AKAP7δ at the cardiac sarcoplasmic reticulum in species other than rat. DNA sequencing identified Human AKAP7δ insertion-deletions (indels) that promote the production of AKAP7γ instead of AKAP7δ. CONCLUSIONS: This AKAP7 molecular evolution study shows that these vital scaffolding proteins developed in ancestral vertebrates and that independent mutations in the AKAP7 genes of rodents and early primates has resulted in the recent formation of AKAP7δ, a splice variant of likely lesser importance in humans than currently described.


Subject(s)
A Kinase Anchor Proteins/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Evolution, Molecular , A Kinase Anchor Proteins/chemistry , Amino Acid Sequence , Animals , Base Sequence , Humans , Membrane Proteins/genetics , Mice , Molecular Sequence Data , Phylogeny , Protein Interaction Domains and Motifs , RNA Splicing , Rats , Sequence Alignment
7.
J Proteomics ; 77: 1-13, 2012 Dec 21.
Article in English | MEDLINE | ID: mdl-22659219

ABSTRACT

There is over-whelming evidence that protein phosphorylations regulate cardiac function and remodeling. A wide variety of protein kinases, e.g., phosphoinositide 3-kinase (PI3K), Akt, GSK-3, TGFß, and PKA, MAPKs, PKC, Erks, and Jaks, as well as phosphatases, e.g., phosphatase I (PP1) and calcineurin, control cardiomyocyte growth and contractility. In the present work, we used global phosphoprotein profiling to identify phosphorylated proteins associated with pressure overload (PO) cardiac hypertrophy and heart failure. Phosphoproteins from hypertrophic and systolic failing hearts from male hypertensive Dahl salt-sensitive rats, trans-aortic banded (TAC), and spontaneously hypertensive heart failure (SHHF) rats were analyzed. Profiling was performed by 2-dimensional difference in gel electrophoresis (2D-DIGE) on phospho-enriched proteins. A total of 25 common phosphoproteins with differences in abundance in (1) the 3 hypertrophic and/or (2) the 2 systolic failure heart models were identified (CI>99%) by matrix assisted laser desorption ionization mass spectrometry (MALDI-MS) and Mascot analysis. Among these were (1) myofilament proteins, including alpha-tropomyosin and myosin regulatory light chain 2, cap Z interacting protein (cap ZIP), and tubulin ß5; (2) mitochondrial proteins, including pyruvate dehydrogenase α, branch chain ketoacid dehydrogenase E1, and mitochondrial creatine kinase; (3) phosphatases, including protein phosphatase 2A and protein phosphatase 1 regulatory subunit; and (4) other proteins including proteosome subunits α type 3 and ß type 7, and eukaryotic translation initiation factor 1A (eIF1A). The results include previously described and novel phosphoproteins in cardiac hypertrophy and systolic failure.


Subject(s)
Hypertension/metabolism , Muscle Proteins/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Phosphoproteins/metabolism , Ventricular Remodeling , Animals , Cardiomegaly/metabolism , Cardiomegaly/pathology , Heart Failure/metabolism , Heart Failure/pathology , Hypertension/pathology , Hypertension/physiopathology , Myocardium/pathology , Myocytes, Cardiac/pathology , Rats , Rats, Inbred SHR , Rats, Wistar , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
8.
Biochim Biophys Acta ; 1822(4): 607-14, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22155497

ABSTRACT

Both histone-acetylations and histone deacetylases have been shown to play a key role in cardiac remodeling. Recently, it has become abundantly clear that many non-histone proteins are modified by post-translational lysine acetylations and that these acetylations regulate protein activity, conformation, and binding. In the present study, non-histone acetylated proteins associated with heart failure were identified. Global screening for lysine acetylated proteins was performed using 2-dimensional gel electrophoresis coupled with immunoblotting with a primary monoclonal anti-acetyl-lysine antibody. Lysine acetylated proteins were compared in two rodent models of hypertensive heart failure, the Dahl salt-sensitive (SS) and spontaneously hypertensive heart failure prone (SHHF) rats with those in corresponding controls, i.e., the Dahl salt-resistant (SR) and W (W) rat strains, respectively. Forty-one and 66 acetylated proteins were detected in SS and SHHF failing hearts, respectively, but either not detected or detected with less abundance in corresponding control hearts. Twelve of these acetylated proteins were common to both models of heart failure. These were identified using matrix-assisted laser desorption/ionization time of flight (MALDI-TOF/TOF) mass spectrometry followed by Mascot Analysis and included mitochondrial enzymes: ATP synthase, long-chain acyl-CoA dehydrogenase, creatine kinase, malate dehydrogenase, and pyruvate dehydrogenase. The abundance of NAD-dependent deacetylase sirtuin-3 (Sirt3), a mitochondrial deacetylase was reduced in SS and SHHF failing hearts. This is the first description of non-histone protein acetylations associated with heart failure and raises the prospect that acetylations of mitochondrial proteins linked to reduced Sirt3 mediate, in part, metabolic changes in heart failure.


Subject(s)
Heart Failure/metabolism , Histones/metabolism , Lysine/metabolism , Muscle Proteins/metabolism , Acetylation , Animals , Muscle Proteins/chemistry , Rats , Rats, Inbred SHR , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
9.
J Exp Biol ; 214(Pt 16): 2709-17, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21795567

ABSTRACT

In mammals, natriuretic peptides (NPs) lower blood pressure, reduce blood volume and broadly inhibit cardiovascular remodeling. NPs are often referred to as cardiac hormones, though they also have integral roles in regulating vascular tone, endothelial remodeling and inhibiting vascular smooth muscle cell hypertrophy. Two NPs [atrial (ANP) and C-type (CNP)] have been identified as endogenous constituents in the vasculature of mammals, though such a phenomenon has not previously been described in fishes. Here we describe the endogenous production of B-type NP (BNP) and CNP in multiple blood vessels of the rainbow trout. Western blot analysis showed pro-BNP and pro-CNP production in the efferent branchial artery, celiacomesenteric artery, ventral aorta and anterior cardinal vein. The detection of pro-BNP and pro-CNP was also supported by MALDI-TOF mass spectrometry analysis of NP-enriched tissue extracts. Although vascular pro-peptide levels of BNP and CNP were quantitatively quite comparable to those found in reference tissues (the atrium for BNP and brain for CNP), mRNA levels of these NPs in the vasculature were greatly reduced as determined by quantitative PCR. When the evolutionarily conserved vascular NP (CNP) was infused into un-anesthetized trout, it reduced central venous pressure and mean circulatory filling pressure. CNP also decreased cardiac output via a reduction in preload. The presence of endogenous NP production in the trout vasculature and potent in vivo hypotensive effects further support the numerous functional similarities between teleost and mammalian NP systems.


Subject(s)
Blood Vessels/metabolism , Natriuretic Peptide, Brain/metabolism , Natriuretic Peptide, C-Type/metabolism , Oncorhynchus mykiss/metabolism , Animals , Base Sequence , Female , Gene Expression Regulation , Heart Function Tests , Male , Molecular Sequence Data , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, C-Type/genetics , Organ Specificity/genetics , Proteomics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Vascular Capacitance
10.
BMC Evol Biol ; 10: 247, 2010 Aug 11.
Article in English | MEDLINE | ID: mdl-20701803

ABSTRACT

BACKGROUND: Cyclic nucleotide phosphodiesterases (PDEs) hydrolyze the intracellular second messengers: cyclic adenosine monophosphate (cAMP) and cyclic guanine monophosphate (cGMP). The cAMP-specific PDE family 4 (PDE4) is widely expressed in vertebrates. Each of the four PDE4 gene isoforms (PDE4 A-D) undergo extensive alternative splicing via alternative transcription initiation sites, producing unique amino termini and yielding multiple splice variant forms from each gene isoform termed long, short, super-short and truncated super-short. Many species across the vertebrate lineage contain multiple splice variants of each gene type, which are characterized by length and amino termini. RESULTS: A phylogenetic approach was used to visualize splice variant form genesis and identify conserved splice variants (genome conservation with EST support) across the vertebrate taxa. Bayesian and maximum likelihood phylogenetic inference indicated PDE4 gene duplication occurred at the base of the vertebrate lineage and reveals additional gene duplications specific to the teleost lineage. Phylogenetic inference and PDE4 splice variant presence, or absence as determined by EST screens, were further supported by the genomic analysis of select vertebrate taxa. Two conserved PDE4 long form splice variants were found in each of the PDE4A, PDE4B, and PDE4C genes, and eight conserved long forms from the PDE4 D gene. Conserved short and super-short splice variants were found from each of the PDE4A, PDE4B, and PDE4 D genes, while truncated super-short variants were found from the PDE4C and PDE4 D genes. PDE4 long form splice variants were found in all taxa sampled (invertebrate through mammals); short, super-short, and truncated super-short are detected primarily in tetrapods and mammals, indicating an increasing complexity in both alternative splicing and cAMP metabolism through vertebrate evolution. CONCLUSIONS: There was a progressive independent incorporation of multiple PDE4 splice variant forms and amino termini, increasing PDE4 proteome complexity from primitive vertebrates to humans. While PDE4 gene isoform duplicates with limited alternative splicing were found in teleosts, an expansion of both PDE4 splice variant forms, and alternatively spliced amino termini predominantly occurs in mammals. Since amino termini have been linked to intracellular targeting of the PDE4 enzymes, the conservation of amino termini in PDE4 splice variants in evolution highlights the importance of compartmentalization of PDE4-mediated cAMP hydrolysis.


Subject(s)
Alternative Splicing , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Evolution, Molecular , Animals , Bayes Theorem , Conserved Sequence , Exons , Gene Duplication , Humans , Isoenzymes/genetics , Likelihood Functions , Multigene Family , Phylogeny , Promoter Regions, Genetic , Protein Interaction Domains and Motifs , Sequence Analysis, DNA
11.
J Exp Biol ; 212(18): 2991-7, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19717682

ABSTRACT

Natriuretic peptides (NPs) and their receptors (NPRs) comprise an evolutionarily conserved signaling system with profound physiological effects on vertebrate renal and cardiovascular systems. Some NPs (ANP, BNP and VNP) are primarily of cardiac origin whereas CNP is common in the brain. In mammals, non-traditional sites of NPs synthesis, BNP in brain and CNP in atrium, appear to have complementary actions. In the present study, trout were chronically adapted to freshwater (FW) (a volume-loading, salt-depleting environment), saltwater (SW) (a volume-depleting, salt-loading environment), FW and fed a high-salt diet (FW-HSD) (a volume- and salt-loading regime) or acutely volume depleted or expanded by hemorrhage or infusion with dialyzed plasma to perturb volume homeostasis. The responses of brain and atrial BNP and CNP mRNA, pro-peptide, NPR-A and NPR-B were evaluated using quantitative PCR and western analysis. Brain pro-BNP and NPR-A was increased in FW-HSD trout and decreased in SW trout. Brain pro-CNP was largely unaffected whereas NPR-B mRNA was increased in FW-HSD trout. Atrial CNP, although produced at lower levels than other cardiac NPs, was markedly elevated in chronically (FW-HSD) and acutely volume expanded trout (dialyzed-plasma infusion) whereas decreased in hemorrhaged trout. These findings indicate that non-traditional NP synthesis sites in the trout probably complement the broad hypovolemic and hypotensive actions of traditional (cardiac) NP synthesis sites in response to volume expansion but not to plasma osmolarity. This supports the hypothesis that the piscine and mammalian NP systems are fundamentally similar and appear to protect the heart from volume overload.


Subject(s)
Fresh Water , Natriuretic Peptides/biosynthesis , Oncorhynchus mykiss/physiology , Protein Isoforms/biosynthesis , Seawater , Animals , Brain/metabolism , Fish Proteins/biosynthesis , Fish Proteins/genetics , Natriuretic Peptides/genetics , Protein Isoforms/genetics , Sodium Chloride, Dietary , Tissue Distribution , Water-Electrolyte Balance
12.
Am J Physiol Regul Integr Comp Physiol ; 296(4): R1170-9, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19176886

ABSTRACT

Natriuretic peptides (NPs) are evolutionarily conserved hormones that affect blood pressure and fluid volume through membrane-bound guanylate cyclase (GC)-linked natriuretic peptide receptors-A and -B (NPR-A and NPR-B, respectively) in a variety of vascular, renal, and other tissues. The principal physiological stimulus for cardiac NPs in fish is somewhat debated between two prominent theories: regulation of salt balance (osmoregulatory hypothesis) or prevention of volume expansion (cardioprotective hypothesis). In the present study, we examined atrial and ventricular expression of trout NPs, atrial (ANP), brain (BNP), and ventricular (VNP) using Northern (mRNA), Western (NP pro-hormone), and qPCR (GC-NPR-A and -B mRNA) blot analysis following independent manipulation of plasma salt and volume levels after chronic exposure to freshwater (FW; volume loaded, salt depleted), saltwater (SW; volume depleted, salt loaded), or freshwater trout fed a high-salt diet (FW-HSD; volume and salt loaded). We also measured NP transcriptional response to acute (2 h) volume expansion with dialyzed plasma (VE; 80% blood vol) or volume depletion by hemorrhage (VD, 20% blood volume every 30 min for 2 h) with real-time PCR. In essentially all instances, increased expression of the NP system was associated with FW-HSD or plasma expansion. There were no differences in NP expression between chronically adapted FW and SW fish, and hemorrhage decreased atrial ANP and VNP mRNA. These results indicate that rainbow trout cardiac NPs and cardiovascular GC-NPRs respond principally to volume, not salt overload, and this suggests that the primary function of trout cardiac NP system is to protect the heart.


Subject(s)
Fish Proteins/metabolism , Myocardium/metabolism , Natriuretic Peptides/metabolism , Oncorhynchus mykiss/metabolism , Receptors, Atrial Natriuretic Factor/metabolism , Water-Electrolyte Balance , Adaptation, Physiological , Animals , Atrial Natriuretic Factor/metabolism , Blood Volume , Chlorides/blood , Fish Proteins/genetics , Fresh Water , Natriuretic Peptide, Brain/metabolism , Natriuretic Peptides/genetics , Oncorhynchus mykiss/blood , Protein Precursors/metabolism , RNA, Messenger/metabolism , Receptors, Atrial Natriuretic Factor/genetics , Seawater , Sodium/blood , Sodium Chloride, Dietary/metabolism , Time Factors
13.
Gen Comp Endocrinol ; 157(1): 21-6, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18472099

ABSTRACT

The natriuretic peptide (NP) family is a seemingly ubiquitous sodium and volume reducing endocrine system of predominantly cardiac origin. Members of the NP system include ANP, BNP, CNP, VNP, their guanylate cyclase (GC)-linked receptors (NPR-A and NPR-B), and clearance receptor (NPR-C). Through the activation of their membrane-bound GC receptors, these small peptides modulate cellular functions that affect both salt and water balance. The elucidation of piscine NP sequences, structure, and functions has steadily advanced over the past 15 years spearheaded by research from Dr. Yoshio Takei's laboratory. The development of these homologous NPs has led to extensive research into both the evolutionary and physiological significance of NPs in fishes. One outcome has been the development of two seemingly disparate hypotheses of NP function; a role in salt excretion, the osmoregulatory hypothesis, versus a role in protecting the heart, the cardioprotective hypotheses. In the osmoregulatory hypothesis NPs are released in response to elevated ambient salinity and inhibit drinking and intestinal uptake of salt, thereby effectively reducing plasma sodium levels. In contrast, the cardioprotective theory depicts NPs acting to prevent debilitating cardiodilation from an excess of either venous or arterial pressure through vasodilation and a reduction of blood volume. These seemingly distinct hypotheses may be elements of a more general regulatory system and certainly require further investigation. Undoubtedly their resolution will not only give us a better perspective of the evolutionary basis of the NP system but will provide us with a greater appreciation of salt and water homeostasis in vertebrates.


Subject(s)
Natriuretic Peptides/physiology , Physiology, Comparative/methods , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , Natriuretic Peptides/genetics , Natriuretic Peptides/metabolism , Physiology, Comparative/trends , Receptors, Atrial Natriuretic Factor/antagonists & inhibitors , Receptors, Atrial Natriuretic Factor/metabolism , Receptors, Atrial Natriuretic Factor/physiology , Sequence Homology, Amino Acid , Water-Electrolyte Balance/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...