Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Neurosci Lett ; 812: 137403, 2023 08 24.
Article in English | MEDLINE | ID: mdl-37473795

ABSTRACT

In males, chronic stress enhances dendritic complexity in the amygdala, a region important in emotion regulation. An amygdalar subregion, the basolateral amygdala (BLA), is influenced by the hippocampus and prefrontal cortex to coordinate emotional learning and memory. This study quantified changes in dendritic complexity of BLA stellate neurons ten days after an unpredictable chronic stressor ended in both male and female rats. In addition, dendritic complexity of hippocampal neurons in male rats was assessed at a similar timepoint. Following Golgi processing, stressed male and female rats showed enhanced BLA dendritic complexity; increased arborization occurred near the soma in males and distally in females. As the brain was sampled ten days after chronic stress ended, BLA dendritic hypertrophy persisted in both sexes after the stressor had ended. For the hippocampus, CA3 dendritic complexity was similar for control and stressed males when assessed eight days after stress ended, suggesting that any stress-induced changes had resolved. These results show persistent enhancement of BLA dendritic arborization in both sexes following chronic stress, reveal sex differences in how BLA hypertrophy manifests, and suggest a putative neurobiological substrate by which chronic stress may create a vulnerable phenotype for emotional dysfunction.


Subject(s)
Amygdala , Dendritic Spines , Hippocampus , Hypertrophy , Neurons , Stress, Psychological , Animals , Female , Male , Rats , Amygdala/pathology , Basolateral Nuclear Complex/pathology , Chronic Disease , Dendritic Spines/pathology , Hippocampus/cytology , Hypertrophy/pathology , Neurons/pathology , Stress, Psychological/pathology , Time Factors , Sex Characteristics , Restraint, Physical
3.
Drug Discov Today ; 26(5): 1107-1110, 2021 05.
Article in English | MEDLINE | ID: mdl-33493454

ABSTRACT

We describe 11 best practices for the successful use of artificial intelligence and machine learning in pharmaceutical and biotechnology research at the data, technology and organizational management levels.


Subject(s)
Artificial Intelligence , Biotechnology/methods , Technology, Pharmaceutical/methods , Humans , Machine Learning , Research Design
4.
Angew Chem Int Ed Engl ; 57(39): 12891-12895, 2018 Sep 24.
Article in English | MEDLINE | ID: mdl-30156746

ABSTRACT

A three-step transformation consisting of 1) addition of electrochemically generated iodosulfonium ions to vinylarenes to give (1-aryl-2-iodoethoxy)sulfonium ions, 2) nucleophilic substitution by subsequently added aromatic compounds to give 1,1-diaryl-2-iodoethane, and 3) elimination of HI with a base to give 1,1-diarylethenes was developed. The transformation serves as a powerful metal- and chemical-oxidant-free method for alkenyl C-H/aromatic C-H cross-coupling.

5.
FEBS J ; 280(10): 2271-84, 2013 May.
Article in English | MEDLINE | ID: mdl-23374253

ABSTRACT

Endorepellin, a processed fragment of perlecan protein core, possesses anti-angiogenic activity by antagonizing endothelial cells. Endorepellin contains three laminin G-like (LG) domains and binds simultaneously to vascular endothelial growth factor receptor 2 (VEGFR2) and α2ß1 integrin, resulting in dual receptor antagonism. Treatment of endothelial cells with endorepellin inhibits transcription of VEGFA, the natural ligand for VEGFR2, attenuating the pro-survival and migratory activities of VEGFA/VEGFR2 signaling cascade. Here, we investigated the specific binding site of endorepellin within the ectodomain of VEGFR2. Full-length endorepellin was not capable of displacing VEGFA binding from VEGFR2 and LG3 domain alone did not bind VEGFR2. This suggested different binding mechanisms of the extracellular Ig domains of VEGFR2. Therefore, we hypothesized that endorepellin would bind through its proximal LG1/2 domains to VEGFR2 in a different region than VEGFA. Indeed, we found that LG1/2 did not bind Ig1-3, but did bind with high affinity to Ig3-5, distal to the known VEGFA binding site, i.e. Ig2-3. These results support a role for endorepellin as an allosteric inhibitor of VEGFR2. Moreover, we found that LG1/2 blocked the rapid VEGFA activation of VEGFR2 at Tyr1175 in endothelial cells. In contrast, LG1/2 did not result in actin cytoskeletal disassembly in endothelial cells whereas LG3 alone did induce cytoskeletal collapse. However, LG1/2 did inhibit VEGFA-dependent endothelial migration through fibrillar collagen I. These studies provide a mechanistic understanding of how the different LG domains of endorepellin signal in endothelial cells while serving as a template for protein design of receptor tyrosine kinase antagonists.


Subject(s)
Endothelial Cells/metabolism , Heparan Sulfate Proteoglycans/metabolism , Neovascularization, Physiologic , Peptide Fragments/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Actin Cytoskeleton , Actins/metabolism , Allosteric Regulation , Angiogenesis Inhibitors/metabolism , Binding Sites , Cell Movement , Collagen/metabolism , HEK293 Cells , Heparan Sulfate Proteoglycans/pharmacology , Humans , Integrin alpha2beta1/metabolism , Peptide Fragments/pharmacology , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transcriptional Activation , Transfection , Tyrosine/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
6.
J Biol Chem ; 287(52): 43543-56, 2012 Dec 21.
Article in English | MEDLINE | ID: mdl-23060442

ABSTRACT

Endorepellin, the angiostatic C-terminal domain of the heparan sulfate proteoglycan perlecan, inhibits angiogenesis by simultaneously binding to the α2ß1 integrin and the vascular endothelial growth factor (VEGF) receptor 2 (VEGFR2) on endothelial cells. This interaction triggers the down-regulation of both receptors and the concurrent activation of the tyrosine phosphatase SHP-1, which leads to a signaling cascade resulting in angiostasis. Here, we provide evidence that endorepellin is capable of attenuating both the PI3K/PDK1/Akt/mTOR and the PKC/JNK/AP1 pathways. We show that hypoxia-inducible factor 1α (HIF-1α) transcriptional activity induced by VEGFA was inhibited by endorepellin independent of oxygen concentration and that only a combination of both PI3K and calcineurin inhibitors completely blocked the suppressive activity evoked by endorepellin on HIF1A and VEGFA promoter activity. Moreover, endorepellin inhibited the PKC/JNK/AP1 axis induced by the recruitment of phospholipase γ and attenuated the VEGFA-induced activation of NFAT1, a process dependent on calcineurin activity. Finally, endorepellin inhibited VEGFA-evoked nuclear translocation of NFAT1 and promoted NFAT1 stability. Thus, we provide evidence for a novel downstream signaling axis for an angiostatic fragment and for the key components involved in the dual antagonistic activity of endorepellin, highlighting its potential use as a therapeutic agent.


Subject(s)
Cell Nucleus/metabolism , Heparan Sulfate Proteoglycans/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , MAP Kinase Signaling System/physiology , NFATC Transcription Factors/metabolism , Neovascularization, Physiologic/physiology , Peptide Fragments/metabolism , Transcription, Genetic/physiology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Active Transport, Cell Nucleus/physiology , Animals , Cell Nucleus/genetics , Cells, Cultured , Heparan Sulfate Proteoglycans/genetics , Human Umbilical Vein Endothelial Cells/cytology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , NFATC Transcription Factors/genetics , Peptide Fragments/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Swine , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
7.
Mol Membr Biol ; 29(2): 36-51, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22416964

ABSTRACT

Abstract Small ankyrin-1 is a splice variant of the ANK1 gene that binds to obscurin A. Previous studies have identified electrostatic interactions that contribute to this interaction. In addition, molecular dynamics (MD) simulations predict four hydrophobic residues in a 'hot spot' on the surface of the ankyrin-like repeats of sAnk1, near the charged residues involved in binding. We used site-directed mutagenesis, blot overlays and surface plasmon resonance assays to study the contribution of the hydrophobic residues, V70, F71, I102 and I103, to two different 30-mers of obscurin that bind sAnk1, Obsc6316₋6345 and Obsc6231₋6260. Alanine mutations of each of the hydrophobic residues disrupted binding to the high affinity binding site, Obsc6316₋6345. In contrast, V70A and I102A mutations had no effect on binding to the lower affinity site, Obsc6231₋6260. Alanine mutagenesis of the five hydrophobic residues present in Obsc6316₋6345 showed that V6328, I6332, and V6334 were critical to sAnk1 binding. Individual alanine mutants of the six hydrophobic residues of Obsc6231₋6260 had no effect on binding to sAnk1, although a triple alanine mutant of residues V6233/I6234/I6235 decreased binding. We also examined a model of the Obsc6316₋6345-sAnk1 complex in MD simulations and found I102 of sAnk1 to be within 2.2Šof V6334 of Obsc6316₋6345. In contrast to the I102A mutation, mutating I102 of sAnk1 to other hydrophobic amino acids such as phenylalanine or leucine did not disrupt binding to obscurin. Our results suggest that hydrophobic interactions contribute to the higher affinity of Obsc6316₋6345 for sAnk1 and to the dominant role exhibited by this sequence in binding.


Subject(s)
Ankyrins/chemistry , Ankyrins/metabolism , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/metabolism , Muscle Proteins/chemistry , Muscle Proteins/metabolism , Alanine/genetics , Alanine/metabolism , Amino Acid Sequence , Animals , Ankyrins/genetics , Binding Sites , Hydrophobic and Hydrophilic Interactions , Leucine/genetics , Leucine/metabolism , Molecular Dynamics Simulation , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Surface Plasmon Resonance
8.
J Mol Biol ; 408(2): 321-34, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21333652

ABSTRACT

Small ankyrin 1 (sAnk1; also known as Ank1.5) is an integral protein of the sarcoplasmic reticulum (SR) in skeletal and cardiac muscle cells, where it is thought to bind to the C-terminal region of obscurin, a large modular protein that surrounds the contractile apparatus. Using fusion proteins in vitro, in combination with site-directed mutagenesis and surface plasmon resonance measurements, we previously showed that the binding site on sAnk1 for obscurin consists, in part, of six lysine and arginine residues. Here we show that four charged residues in the high-affinity binding site on obscurin for sAnk1 (between residues 6316 and 6345), consisting of three glutamates and a lysine, are necessary, but not sufficient, for this site on obscurin to bind to sAnk1 with high affinity. We also identify specific complementary mutations in sAnk1 that can partially or completely compensate for the changes in binding caused by charge-switching mutations in obscurin. We used molecular modeling to develop structural models of residues 6322-6339 of obscurin bound to sAnk1. The models, based on a combination of Brownian and molecular dynamics simulations, predict that the binding site on sAnk1 for obscurin is organized as two ankyrin-like repeats, with the last α-helical segment oriented at an angle to nearby helices, allowing lysine 6338 of obscurin to form an ionic interaction with aspartate 111 of sAnk1. This prediction was validated by double-mutant cycle experiments. Our results are consistent with a model in which electrostatic interactions between specific pairs of side chains on obscurin and sAnk1 promote binding and complex formation.


Subject(s)
Ankyrins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Models, Molecular , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Sarcoplasmic Reticulum/metabolism , Ankyrins/chemistry , Ankyrins/genetics , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/genetics , Humans , Maltose-Binding Proteins/genetics , Maltose-Binding Proteins/metabolism , Molecular Dynamics Simulation , Muscle Proteins/chemistry , Muscle Proteins/genetics , Mutagenesis, Site-Directed , Mutation/genetics , Protein Conformation , Protein Serine-Threonine Kinases , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Rho Guanine Nucleotide Exchange Factors , Surface Plasmon Resonance
9.
Biochemistry ; 49(46): 9948-56, 2010 Nov 23.
Article in English | MEDLINE | ID: mdl-20949908

ABSTRACT

Obscurin A, an ∼720 kDa modular protein of striated muscles, binds to small ankyrin 1 (sAnk1, Ank 1.5), an integral protein of the sarcoplasmic reticulum, through two distinct carboxy-terminal sequences, Obsc(6316-6436) and Obsc(6236-6260). We hypothesized that these sequences differ in affinity but that they compete for the same binding site on sAnk1. We show that the sequence within Obsc(6316-6436) that binds to sAnk1 is limited to residues 6316-6345. Comparison of Obsc(6231-6260) to Obsc(6316-6345) reveals that Obsc(6316-6345) binds sAnk1 with an affinity (133 ± 43 nM) comparable to that of the Obsc(6316-6436) fusion protein, whereas Obsc(6231-6260) binds with lower affinity (384 ± 53 nM). Oligopeptides of each sequence compete for binding with both sites at half-maximal inhibitory concentrations consistent with the affinities measured directly. Five of six site-directed mutants of sAnk1 showed similar reductions in binding to each binding site on obscurin, suggesting that they dock to many of the same residues of sAnk1. Circular dichroism (CD) analysis of the synthetic oligopeptides revealed a 2-fold greater α-helical content in Obsc(6316-6346), ∼35%, than Obsc(6231-6260,) ∼17%. Using these data, structural prediction algorithms, and homology modeling, we predict that Obsc(6316-6345) contains a bent α-helix of 12 amino acids, flanked by short disordered regions, and that Obsc(6231-6260) has a short, N-terminal α-helix of 4-5 residues followed by a long disordered region. Our results are consistent with a model in which both sequences of obscurin differ significantly in structure but bind to the ankyrin-like repeat motifs of sAnk1 in a similar though not identical manner.


Subject(s)
Ankyrins/chemistry , Muscle Proteins/chemistry , Algorithms , Amino Acid Sequence , Ankyrins/metabolism , Binding Sites , Cells, Cultured , Circular Dichroism , Molecular Sequence Data , Muscle Proteins/metabolism , Sarcoplasmic Reticulum/metabolism
10.
J Biol Chem ; 282(29): 20948-59, 2007 Jul 20.
Article in English | MEDLINE | ID: mdl-17517897

ABSTRACT

The RNA-binding factor HuR is a ubiquitously expressed member of the Hu protein family that binds and stabilizes mRNAs containing AU-rich elements (AREs). Hu proteins share a common domain organization of two tandemly arrayed RNA recognition motifs (RRMs) near the N terminus, followed by a basic hinge domain and a third RRM near the C terminus. In this study, we engineered recombinant wild-type and mutant HuR proteins lacking affinity tags to characterize their ARE-binding properties. Using combinations of electrophoretic mobility shift and fluorescence anisotropy-based binding assays, we show that HuR can bind ARE substrates as small as 13 nucleotides with low nanomolar affinity, but forms cooperative oligomeric protein complexes on ARE substrates of at least 18 nucleotides in length. Analyses of deletion mutant proteins indicated that RRM3 does not contribute to high affinity recognition of ARE substrates, but is required for cooperative assembly of HuR oligomers on RNA. Finally, the hinge domain between RRM2 and RRM3 contributes significant binding energy to HuR.ARE complex formation in an ARE length-dependent manner. The hinge does not enhance RNA-binding activity by increased ion pair formation despite extensive positive charge within this region, and it does not thermodynamically stabilize protein folding. Together, the results define distinct roles for the HuR hinge and RRM3 domains in formation of cooperative HuR.ARE complexes in solution.


Subject(s)
Antigens, Surface/chemistry , ELAV Proteins/chemistry , RNA, Messenger/chemistry , RNA-Binding Proteins/chemistry , Amino Acid Motifs , Amino Acid Sequence , ELAV-Like Protein 1 , Fluorescence Polarization , Gene Deletion , Genetic Vectors , Humans , Molecular Sequence Data , Nanotechnology , Protein Binding , Protein Folding , Protein Structure, Tertiary , RNA, Messenger/metabolism , Sequence Homology, Amino Acid
11.
Phys Ther ; 82(3): 257-63, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11869154

ABSTRACT

BACKGROUND AND PURPOSE: Physiological effects of therapeutic ultrasound (US) are dependent on the intensity and duration of application. The purpose of this study was to test US machines used in clinical settings for proper calibration of time and power output. METHODS: Measurements of power output and timer accuracy were obtained from 83 US units in clinical use. The machines were tested at 4 intensity settings (0.5, 1.0, 1.5, and 2.0 W/cm2) using a continuous waveform and a 1-MHz frequency. The measured intensities were converted to percentages of error and compared with the +/-20% standard. RESULTS: Of the machines tested, 32 (39%) were outside the calibration standard for at least one output setting. Of these machines, 15 (18%) were above the +20% standard, and 17 (21%) were below the -20% standard for at least one output setting. Of the 32 machines outside the standard, 26 (31%) were outside the standard for 2 or more settings, and 3 (4%) produced no output at any of the settings. Of the mechanical timers tested, 7 (28%) were outside of the +/-10% standard for timer accuracy at the 5-minute interval, and 6 (24%) were outside of the standard at the 10-minute interval. All digital timers tested were within the standard. DISCUSSION AND CONCLUSION: More than one third of machines tested in this study were outside the standard for power output, and approximately one fourth of the mechanical timers were outside the standard. Therefore, further improvements in the accuracy of US machine calibration are needed.


Subject(s)
Physical Therapy Specialty , Ultrasonic Therapy/standards , Equipment Design , Humans , Pilot Projects , Surveys and Questionnaires , Texas , Ultrasonic Therapy/instrumentation
SELECTION OF CITATIONS
SEARCH DETAIL
...