Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Toxins (Basel) ; 13(1)2020 Dec 24.
Article in English | MEDLINE | ID: mdl-33374102

ABSTRACT

Enterohemorrhagic Escherichia coli (EHEC) infections can cause EHEC-associated hemolytic uremic syndrome (eHUS) via its main virulent factor, Shiga toxins (Stxs). Complement has been reported to be involved in the progression of eHUS. The aim of this study was to investigate the interactions of the most effective subtype of the toxin, Stx2a, with pivotal complement proteins C3b and C5. The study further examined the effect of Stx2a stimulation on the transcription and synthesis of these complement proteins in human target cell lines. Binding of Stx2a to C3b and C5 was evaluated by ELISA. Kidney and gut cell lines (HK-2 and HCT-8) were stimulated with varied concentrations of Stx2a. Subsequent evaluation of complement gene transcription was studied by real-time PCR (qPCR), and ELISAs and Western blots were performed to examine protein synthesis of C3 and C5 in supernatants and lysates of stimulated HK-2 cells. Stx2a showed a specific binding to C3b and C5. Gene transcription of C3 and C5 was upregulated with increasing concentrations of Stx2a in both cell lines, but protein synthesis was not. This study demonstrates the binding of Stx2a to complement proteins C3b and C5, which could potentially be involved in regulating complement during eHUS infection, supporting further investigations into elucidating the role of complement in eHUS pathogenesis.


Subject(s)
Complement C3b/chemistry , Complement C5/chemistry , Gene Expression Regulation/drug effects , Shiga Toxin/chemistry , Shiga Toxin/pharmacology , Cell Line , Cell Survival , Humans , Protein Binding , Up-Regulation/drug effects
2.
J Am Soc Mass Spectrom ; 31(9): 1783-1802, 2020 Sep 02.
Article in English | MEDLINE | ID: mdl-32812765

ABSTRACT

The Consortium for Top-Down Proteomics (www.topdownproteomics.org) launched the present study to assess the current state of top-down mass spectrometry (TD MS) and middle-down mass spectrometry (MD MS) for characterizing monoclonal antibody (mAb) primary structures, including their modifications. To meet the needs of the rapidly growing therapeutic antibody market, it is important to develop analytical strategies to characterize the heterogeneity of a therapeutic product's primary structure accurately and reproducibly. The major objective of the present study is to determine whether current TD/MD MS technologies and protocols can add value to the more commonly employed bottom-up (BU) approaches with regard to confirming protein integrity, sequencing variable domains, avoiding artifacts, and revealing modifications and their locations. We also aim to gather information on the common TD/MD MS methods and practices in the field. A panel of three mAbs was selected and centrally provided to 20 laboratories worldwide for the analysis: Sigma mAb standard (SiLuLite), NIST mAb standard, and the therapeutic mAb Herceptin (trastuzumab). Various MS instrument platforms and ion dissociation techniques were employed. The present study confirms that TD/MD MS tools are available in laboratories worldwide and provide complementary information to the BU approach that can be crucial for comprehensive mAb characterization. The current limitations, as well as possible solutions to overcome them, are also outlined. A primary limitation revealed by the results of the present study is that the expert knowledge in both experiment and data analysis is indispensable to practice TD/MD MS.


Subject(s)
Antibodies, Monoclonal , Mass Spectrometry/methods , Proteomics/methods , Animals , Antibodies, Monoclonal/analysis , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Complementarity Determining Regions/analysis , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/genetics , Humans , Mice
3.
Front Microbiol ; 11: 914, 2020.
Article in English | MEDLINE | ID: mdl-32499768

ABSTRACT

Developing cultivation methods that yield chemically and isotopically defined fatty acid (FA) compositions within bacterial cytoplasmic membranes establishes an in vivo experimental platform to study membrane biophysics and cell membrane regulation using novel approaches. Yet before fully realizing the potential of this method, it is prudent to understand the systemic changes in cells induced by the labeling procedure itself. In this work, analysis of cellular membrane compositions was paired with proteomics to assess how the proteome changes in response to the directed incorporation of exogenous FAs into the membrane of Bacillus subtilis. Key findings from this analysis include an alteration in lipid headgroup distribution, with an increase in phosphatidylglycerol lipids and decrease in phosphatidylethanolamine lipids, possibly providing a fluidizing effect on the cell membrane in response to the induced change in membrane composition. Changes in the abundance of enzymes involved in FA biosynthesis and degradation are observed; along with changes in abundance of cell wall enzymes and isoprenoid lipid production. The observed changes may influence membrane organization, and indeed the well-known lipid raft-associated protein flotillin was found to be substantially down-regulated in the labeled cells - as was the actin-like protein MreB. Taken as a whole, this study provides a greater depth of understanding for this important cell membrane experimental platform and presents a number of new connections to be explored in regard to modulating cell membrane FA composition and its effects on lipid headgroup and raft/cytoskeletal associated proteins.

4.
J Mass Spectrom ; 55(7): e4446, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31652378

ABSTRACT

The identification of peptides and proteins from tandem mass spectra is a difficult task and multiple tools have been developed to aid this identification. We present a new method called quantum chemical mass spectrometry for materials science (QCMS2 ), which is based on quantum chemical calculations of bond orders, reaction, and transition-state energies at the DFT/B3LYP/6-311+G* level of theory. The method was used to describe the fragmentation pathways of five X-His-Ser tripeptides with X = Asn, Asp, Glu, Ser, and Trp, thereby focusing on the influence of the side chain and inter-side-chain interactions on the fragmentation. The main features in the mass spectra of the five tripeptides were correctly reproduced, and a number of fragments were assigned to fragmentations involving the side chain and the influence of inter-side-chain interactions. Product ion spectra were recorded to evaluate the capabilities and limitations of QCMS2 and a number of conventional tools.


Subject(s)
Peptide Fragments/chemistry , Peptides/chemistry , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry/methods , Hydrogen Bonding , Models, Molecular , Molecular Structure
5.
Nat Commun ; 10(1): 3166, 2019 07 18.
Article in English | MEDLINE | ID: mdl-31320618

ABSTRACT

Aurora kinases B and C (AURKB/AURKC) are activated by binding to the C-terminal domain of INCENP. Full activation requires phosphorylation of two serine residues of INCENP that are conserved through evolution, although the mechanism of this activation has not been explained. Here we present crystal structures of the fully active complex of AURKC bound to INCENP, consisting of phosphorylated, activated, AURKC and INCENP phosphorylated on its TSS motif, revealing the structural and biochemical mechanism of synergistic activation of AURKC:INCENP. The structures show that TSS motif phosphorylation stabilises the kinase activation loop of AURKC. The TSS motif phosphorylations alter the substrate-binding surface consistent with a mechanism of altered kinase substrate selectivity and stabilisation of the protein complex against unfolding. We also analyse the binding of the most specific available AURKB inhibitor, BRD-7880, and demonstrate that the well-known Aurora kinase inhibitor VX-680 disrupts binding of the phosphorylated INCENP TSS motif.


Subject(s)
Aurora Kinase B/metabolism , Aurora Kinase C/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Crystallography, X-Ray , Enzyme Activation , Humans , Phosphorylation , Piperazines/pharmacology , Protein Binding/drug effects , Protein Domains , Protein Folding , Serine/metabolism
6.
Cell Microbiol ; 21(5): e13000, 2019 05.
Article in English | MEDLINE | ID: mdl-30578712

ABSTRACT

Hemolytic uremic syndrome (eHUS) is a severe complication of human infections with Shiga toxins (Stxs)-producing Escherichia coli. A key step in the pathogenesis of eHUS is the interaction of Stxs with blood components before the targeting of renal endothelial cells. Here, we show that a single proteolytic cleavage in the Stx2a A-subunit, resulting into two fragments (A1 and A2) linked by a disulfide bridge (cleaved Stx2a), dictates different binding abilities. Uncleaved Stx2a was confirmed to bind to human neutrophils and to trigger leukocyte/platelet aggregate formation, whereas cleaved Stx2a was ineffective. Conversely, binding of complement factor H was confirmed for cleaved Stx2a and not for uncleaved Stx2a. It is worth noting that uncleaved and cleaved Stx2a showed no differences in cytotoxicity for Vero cells or Raji cells, structural conformation, and contaminating endotoxin. These results have been obtained by comparing two Stx2a batches, purified in different laboratories by using different protocols, termed Stx2a(cl; cleaved toxin, Innsbruck) and Stx2a(uncl; uncleaved toxin, Bologna). Stx2a(uncl) behaved as Stx2a(cl) after mild trypsin treatment. In this light, previous controversial results obtained with purified Stx2a has to be critically re-evaluated; furthermore, characterisation of the structure of circulating Stx2a is mandatory to understand eHUS-pathogenesis and to develop therapeutic approaches.


Subject(s)
Escherichia coli/chemistry , Shiga Toxin 2/chemistry , Shiga Toxin 2/metabolism , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , Chlorocebus aethiops , Circular Dichroism , Complement Factor H/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Fluorescence , Humans , Leukocytes/drug effects , Leukocytes/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Protein Binding , Protein Conformation , Shiga Toxin 2/genetics , Trihexosylceramides/metabolism , Trypsin , Vero Cells
7.
Int J Med Microbiol ; 308(7): 969-976, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30064820

ABSTRACT

Escherichia coli-induced hemolytic uremic syndrome (eHUS) is a life-threatening complication of infection with Shiga toxin (Stx), in particular Stx2a-producing Escherichia coli. Enhanced coagulation activation with formation of microthrombi seems to be a key event in development of eHUS. Platelet activation has been postulated as a possible, but controversially debated mechanism. The present study investigated the effect of Stx2a on plasmatic coagulation and platelets. Binding studies were initially performed with ELISA and co-immunoprecipitation and supported by quartz crystal microbalance with dissipation monitoring (QCM-D). Antithrombin (AT) activity was measured using the automated BCS XP® system. ROTEM® was used for functional coagulation testing. Platelet binding and activation was studied with FACS and light-transmission aggregometry. We found binding of Stx2a to AT, an important inhibitor of blood coagulation, but only a mild albeit significant reduction of AT activity against FXa in the presence of Stx2a. QCM-D analysis also showed binding of Stx2a to heparin and an impaired binding of AT to Stx2a-bound heparin. ROTEM® using Stx2a-treated platelet-poor plasma revealed a significant, but only moderate shortening of clotting time. Neither binding nor activation of platelets by Stx2a could be demonstrated. In summary, data of this study suggest that Stx2a binds to AT, but does not induce major effects on plasmatic coagulation. In addition, no interaction with platelets occurred. The well-known non-beneficial administration of heparin in eHUS patients could be explained by the interaction of Stx2a with heparin.


Subject(s)
Antithrombins/metabolism , Blood Coagulation/physiology , Heparin/metabolism , Platelet Aggregation/immunology , Shiga Toxin 2/metabolism , Blood Platelets/immunology , Hemolytic-Uremic Syndrome/microbiology , Humans , Protein Binding/physiology , Shiga-Toxigenic Escherichia coli/pathogenicity
8.
J Sep Sci ; 41(1): 125-144, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28990739

ABSTRACT

The characterization of biotherapeutics represents a major analytical challenge. This review discusses the current state-of-the-art in analytical technologies to profile biopharma products under native conditions, i.e., the protein three dimensional conformation is maintained during liquid chromatographic analysis. Native liquid-chromatographic modes that are discussed include aqueous size-exclusion chromatography, hydrophobic interaction chromatography, and ion-exchange chromatography. Infusion conditions and the possibilities and limitations to hyphenate native liquid chromatography to mass spectrometry are discussed. Furthermore, the applicability of native liquid-chromatography methods and intact mass spectrometry analysis for the characterization of monoclonal antibodies and antibody-drug conjugates is discussed.


Subject(s)
Antibodies, Monoclonal/analysis , Biological Products/analysis , Chemistry Techniques, Analytical , Chromatography, High Pressure Liquid/trends , Mass Spectrometry/trends , Proteins/analysis , Animals , Chromatography, Liquid , Humans , Hydrogen-Ion Concentration , Protein Denaturation , Protein Folding , Shear Strength , Spectrometry, Mass, Electrospray Ionization , Stress, Mechanical
9.
Front Microbiol ; 9: 3319, 2018.
Article in English | MEDLINE | ID: mdl-30697200

ABSTRACT

Complement is a tightly controlled arm of the innate immune system, facilitating phagocytosis and killing of invading pathogens. Factor H (FH) is the main fluid-phase inhibitor of the alternative pathway. Many pathogens can hijack FH from the host and protect themselves from complement-dependent killing. Candida albicans is a clinically important opportunistic yeast, expressing different FH binding molecules on its cell surface, which allow complement evasion. One such FH binding molecule is the transmembrane protein "High affinity glucose transporter 1" (Hgt1p), involved in glucose metabolism. This study demonstrated that Hgt1p transcription and expression is induced and highest at the low, but physiological glucose concentration of 0.1%. Thus, this concentration was used throughout the study. We also demonstrated the transport of Hgt1p to the fungal cell wall surface by vesicle trafficking and its release by exosomes containing Hgt1p integrated in the vesicular membrane. We corroborated Hgt1p as FH binding molecule. A polyclonal anti-Hgt1p antibody was created which interfered with the binding of FH, present in normal human serum to the fungal cell wall. A chimeric molecule consisting of FH domains 6 and 7 fused to human IgG1 Fc (FH6.7/Fc) even more comprehensively blocked FH binding, likely because FH6.7/Fc diverted FH away from fungal FH ligands other than Hgt1p. Reduced FH binding to the yeast was associated with a concomitant increase in C3b/iC3b deposition and resulted in significantly increased in vitro phagocytosis and killing by human neutrophils. In conclusion, Hgt1p also exhibits non-canonical functions such as binding FH after its export to the cell wall. Blocking Hgt1p-FH interactions may represent a tool to enhance complement activation on the fungal surface to promote phagocytosis and killing of C. albicans.

10.
J Phys Chem Lett ; 8(17): 4214-4217, 2017 Sep 07.
Article in English | MEDLINE | ID: mdl-28825491

ABSTRACT

Lipid extracts are an excellent choice of model biomembrane; however at present, there are no commercially available lipid extracts or computational models that mimic microbial membranes containing the branched-chain fatty acids found in many pathogenic and industrially relevant bacteria. We advance the extract of Bacillus subtilis as a standard model for these diverse systems, providing a detailed experimental description and equilibrated atomistic bilayer model included as Supporting Information to this Letter and at ( http://cmb.ornl.gov/members/cheng ). The development and validation of this model represents an advance that enables more realistic simulations and experiments on bacterial membranes and reconstituted bacterial membrane proteins.


Subject(s)
Bacillus subtilis , Cell Membrane/physiology , Membrane Proteins/chemistry , Models, Biological , Bacterial Proteins , Fatty Acids , Lipid Bilayers , Membrane Lipids
11.
PLoS Biol ; 15(5): e2002214, 2017 05.
Article in English | MEDLINE | ID: mdl-28542493

ABSTRACT

Examining the fundamental structure and processes of living cells at the nanoscale poses a unique analytical challenge, as cells are dynamic, chemically diverse, and fragile. A case in point is the cell membrane, which is too small to be seen directly with optical microscopy and provides little observational contrast for other methods. As a consequence, nanoscale characterization of the membrane has been performed ex vivo or in the presence of exogenous labels used to enhance contrast and impart specificity. Here, we introduce an isotopic labeling strategy in the gram-positive bacterium Bacillus subtilis to investigate the nanoscale structure and organization of its plasma membrane in vivo. Through genetic and chemical manipulation of the organism, we labeled the cell and its membrane independently with specific amounts of hydrogen (H) and deuterium (D). These isotopes have different neutron scattering properties without altering the chemical composition of the cells. From neutron scattering spectra, we confirmed that the B. subtilis cell membrane is lamellar and determined that its average hydrophobic thickness is 24.3 ± 0.9 Ångstroms (Å). Furthermore, by creating neutron contrast within the plane of the membrane using a mixture of H- and D-fatty acids, we detected lateral features smaller than 40 nm that are consistent with the notion of lipid rafts. These experiments-performed under biologically relevant conditions-answer long-standing questions in membrane biology and illustrate a fundamentally new approach for systematic in vivo investigations of cell membrane structure.


Subject(s)
Bacillus subtilis/metabolism , Cell Membrane/metabolism , Fatty Acids/metabolism , Lipid Bilayers/metabolism , Membrane Microdomains/metabolism , Models, Biological , Algorithms , Bacillus subtilis/chemistry , Bacillus subtilis/drug effects , Bacillus subtilis/growth & development , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Membrane/chemistry , Cell Membrane/drug effects , Cerulenin/pharmacology , Deuterium , Enoyl-CoA Hydratase/genetics , Enoyl-CoA Hydratase/metabolism , Fatty Acid Synthesis Inhibitors/pharmacology , Fatty Acids/chemistry , Gene Deletion , Hydrophobic and Hydrophilic Interactions , Lipid Bilayers/chemistry , Membrane Microdomains/chemistry , Membrane Microdomains/drug effects , Microbial Viability/drug effects , Neutron Diffraction , Palmitic Acids/chemistry , Palmitic Acids/metabolism , Scattering, Small Angle , Stereoisomerism
12.
Nat Chem Biol ; 13(6): 640-646, 2017 06.
Article in English | MEDLINE | ID: mdl-28369041

ABSTRACT

Toxin-antitoxin (TA) loci are prevalent in bacterial genomes. They are suggested to play a central role in dormancy and persister states. Under normal growth conditions, TA toxins are neutralized by their cognate antitoxins, and under stress conditions, toxins are freed and inhibit essential cellular processes using a variety of mechanisms. Here we characterize ataR-ataT, a novel TA system, from enterohemorrhagic Escherichia coli. We show that the toxin AtaT is a GNAT family enzyme that transfers an acetyl group from acetyl coenzyme A to the amine group of the methionyl aminoacyl moiety of initiator tRNA. AtaT specifically modifies Met-tRNAfMet, but no other aminoacyl-tRNAs, including the elongator Met-tRNAMet. We demonstrate that once acetylated, AcMet-tRNAfMet fails to interact with initiation factor-2 (IF2), resulting in disruption of the translation initiation complex. This work reveals a new mechanism of translation inhibition and confirms Met-tRNAfMet as a prime target to efficiently block cell growth.


Subject(s)
Amino-Acid N-Acetyltransferase/metabolism , Escherichia coli , Gene Expression Regulation/genetics , RNA, Transfer, Met/metabolism , Acetylation , Electrophoresis, Gel, Two-Dimensional , Models, Biological , Protein Biosynthesis
13.
Org Biomol Chem ; 14(31): 7544-51, 2016 Aug 21.
Article in English | MEDLINE | ID: mdl-27431745

ABSTRACT

Recently, bioorthogonal chemistry based on the Inverse Electron-Demand Diels-Alder (IEDDA) cycloaddition between 1,2,4,5-tetrazines and trans-cyclooctene (TCO) analogues added an interesting dimension to molecular imaging. Until now, antibodies (Abs) were tagged with TCO and after pretargeting they were reacted with tetrazines substituted with reporters. However, TCO tags have the tendency to degrade under physiological conditions, and due to their hydrophobic nature are buried within the protein. This results in loss of reactivity and a low Ab functional loading. To circumvent these problems, we report for the first time an approach in which tetrazines are used as tags for antibody (Ab) modification, and TCO as the imaging agent. We developed a new Ab-tetrazine conjugate, which displays a high functional loading, good stability and reactivity. We utilized this immunoconjugate for live-cell imaging together with novel TCO probes, resulting in selective and rapid labeling of SKOV-3 cells. Our approach may be useful for in vivo pretargeted imaging.

14.
J Biol Chem ; 290(7): 4178-91, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25505247

ABSTRACT

The multisubunit cullin RING E3 ubiquitin ligases (CRLs) target post-translationally modified substrates for ubiquitination and proteasomal degradation. The suppressors of cytokine signaling (SOCS) proteins play important roles in inflammatory processes, diabetes, and cancer and therefore represent attractive targets for therapeutic intervention. The SOCS proteins, among their other functions, serve as substrate receptors of CRL5 complexes. A member of the CRL family, SOCS2-EloBC-Cul5-Rbx2 (CRL5(SOCS2)), binds phosphorylated growth hormone receptor as its main substrate. Here, we demonstrate that the components of CRL5(SOCS2) can be specifically pulled from K562 human cell lysates using beads decorated with phosphorylated growth hormone receptor peptides. Subsequently, SOCS2-EloBC and full-length Cul5-Rbx2, recombinantly expressed in Escherichia coli and in Sf21 insect cells, respectively, were used to reconstitute neddylated and unneddylated CRL5(SOCS2) complexes in vitro. Finally, diverse biophysical methods were employed to study the assembly and interactions within the complexes. Unlike other E3 ligases, CRL5(SOCS2) was found to exist in a monomeric state as confirmed by size exclusion chromatography with inline multiangle static light scattering and native MS. Affinities of the protein-protein interactions within the multisubunit complex were measured by isothermal titration calorimetry. A structural model for full-size neddylated and unneddylated CRL5(SOCS2) complexes is supported by traveling wave ion mobility mass spectrometry data.


Subject(s)
Cullin Proteins/metabolism , Protein Conformation , Suppressor of Cytokine Signaling Proteins/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitins/metabolism , Cullin Proteins/chemistry , Cullin Proteins/genetics , Elongin , Humans , K562 Cells , Mass Spectrometry , Models, Molecular , NEDD8 Protein , Protein Binding , Suppressor of Cytokine Signaling Proteins/chemistry , Suppressor of Cytokine Signaling Proteins/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitination , Ubiquitins/chemistry , Ubiquitins/genetics
15.
Development ; 141(2): 335-45, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24353062

ABSTRACT

Organ development is a highly regulated process involving the coordinated proliferation and differentiation of diverse cellular populations. The pathways regulating cell proliferation and their effects on organ growth are complex and for many organs incompletely understood. In all vertebrate species, the cardiac natriuretic peptides (ANP and BNP) are produced by cardiomyocytes in the developing heart. However, their role during cardiogenesis is not defined. Using the embryonic zebrafish and neonatal mammalian cardiomyocytes we explored the natriuretic peptide signaling network during myocardial development. We observed that the cardiac natriuretic peptides ANP and BNP and the guanylate cyclase-linked natriuretic peptide receptors Npr1 and Npr2 are functionally redundant during early cardiovascular development. In addition, we demonstrate that low levels of the natriuretic peptides preferentially activate Npr3, a receptor with Gi activator sequences, and increase cardiomyocyte proliferation through inhibition of adenylate cyclase. Conversely, high concentrations of natriuretic peptides reduce cardiomyocyte proliferation through activation of the particulate guanylate cyclase-linked natriuretic peptide receptors Npr1 and Npr2, and activation of protein kinase G. These data link the cardiac natriuretic peptides in a complex hierarchy modulating cardiomyocyte numbers during development through opposing effects on cardiomyocyte proliferation mediated through distinct cyclic nucleotide signaling pathways.


Subject(s)
Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Receptors, Atrial Natriuretic Factor/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Cell Proliferation , Cyclic AMP/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Gene Knockdown Techniques , Heart/embryology , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, Brain/metabolism , Receptors, Atrial Natriuretic Factor/antagonists & inhibitors , Receptors, Atrial Natriuretic Factor/genetics , Signal Transduction , Zebrafish/genetics , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...