Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Cell Physiol Biochem ; 53(2): 400-412, 2019.
Article in English | MEDLINE | ID: mdl-31403270

ABSTRACT

BACKGROUND/AIMS: Mutations in ABCA4 cause Stargardt macular degeneration, which invariably ends in legal blindness. We studied two common mutants, A1038V (in NBD1) and G1961E (in NBD2), with the purpose of exploring how they interact with the cell's quality control mechanism. The study was designed to determine how these mutants can be rescued. METHODS: We expressed wt and mutant ABCA4 in HEK293 cells and studied the effect of the mutations on trafficking and processing and the ability of correctors to rescue them. We used a combination of western blotting, confocal microscopy and surface biotinylation coupled with pulldown of plasma membrane proteins. RESULTS: G1961E is sensitive to inhibitors of the aggresome, tubacin and the lysosome, bafilomycin A. Both mutants cause a reduction in heat shock protein, Hsp27. Incubation of HEK293 cells expressing the mutants with VX-809, an FDA approved drug for the treatment of cystic fibrosis, increased the levels of A1038V and G1961E by 2- to 3-fold. Importantly, VX-809 increased the levels of both mutants at the plasma membrane suggesting that trafficking had been restored. Transfecting additional Hsp27 to the cells also increased the steady state levels of both mutants. However, in combination with VX-809 the addition of Hsp27 caused a dramatic increase in the protein expression particularly in the G1961 mutant which increased approximately 5-fold. CONCLUSION: Our results provide a new mechanism for the rescue of ABCA4 trafficking mutants based on the restoration of Hsp27. Our results provide a pathway for the treatment of Stargardt disease.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Aminopyridines/pharmacology , Benzodioxoles/pharmacology , ATP-Binding Cassette Transporters/genetics , Aminopyridines/therapeutic use , Anilides/pharmacology , Benzodioxoles/therapeutic use , Cell Membrane/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Gene Expression Regulation/drug effects , HEK293 Cells , HSP27 Heat-Shock Proteins/metabolism , Humans , Hydroxamic Acids/pharmacology , Leupeptins/pharmacology , Lysosomes/metabolism , Macular Degeneration/congenital , Macular Degeneration/drug therapy , Macular Degeneration/metabolism , Macular Degeneration/pathology , Mutation , Protein Transport/drug effects , Stargardt Disease
2.
Cell Physiol Biochem ; 52(6): 1267-1279, 2019.
Article in English | MEDLINE | ID: mdl-31026390

ABSTRACT

BACKGROUND/AIMS: Because of the small size of adeno-associated virus, AAV, the cystic fibrosis conductance regulator, CFTR, cDNA is too large to fit within AAV and must be truncated. We report here on two truncated versions of CFTR, which, when inserted into AAV1 and used to infect airway cells, rescue F508-del CFTR via transcomplementation. The purpose of this study is to shed light on where in the cell transcomplementation occurs and how it results in close association between the endogenous F508-del and truncated CFTR. METHODS: We treated CF airway cells (CFBE41o-) with AAV2/1 (AAV2 inverted terminal repeats/AAV1 capsid) containing truncated forms of CFTR, ∆264 and ∆27-264 CFTR, who can restore the function of F508-del by transcomplementation. We addressed the aims of the study using a combination of confocal microscopy and short circuit currents measurements. For the latter, CF bronchial epithelial cells (CFBE) were grown on permeable supports. RESULTS: We show that both F508del and the truncation mutants colocalize in the ER and that both the rescued F508-del and the transcomplementing mutants reach the plasma membrane together. There was significant fluorescence resonance energy transfer (FRET) between F508-del and the transcomplementing mutants within the endoplasmic reticulum (ER), suggesting that transcomplementation occurs through a bimolecular interaction. We found that transcomplementation could increase the Isc in CFBE41o- cells stably expressing additional wt-CFTR or F508-del and in parental CFBE41o- cells expressing endogenous levels of F508-del. CONCLUSION: We conclude that the functional rescue of F508-del by transcomplementation occurs via a bimolecular interaction that most likely begins in the ER and continues at the plasma membrane. These results come at an opportune time for developing a gene therapy for CF and offer new treatment options for a wide range of CF patients.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/genetics , Dependovirus/genetics , Endoplasmic Reticulum/genetics , Cell Line , Cystic Fibrosis/therapy , Genetic Therapy , Humans , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , Sequence Deletion , Transfection
3.
Cell Physiol Biochem ; 51(3): 1489-1499, 2018.
Article in English | MEDLINE | ID: mdl-30485852

ABSTRACT

BACKGROUND/AIMS: Cystic fibrosis (CF) is a lethal recessive disorder caused by mutations in the CF transmembrane conductance regulator (CFTR). ΔF508, the most common mutation, is a misfolded protein that is retained in the endoplasmic reticulum and degraded, precluding delivery to the cell surface [1]. METHODS: Here we use a combination of western blotting, immunoprecipitation, and short circuit current techniques combined with confocal microscopy to address whether the SNARE attachment protein, STX8 plays a role in ΔF508's processing and movement out of the ER. RESULTS: Although the SNARE protein STX8 is thought to be functionally related and primarily localized to early endosomes, we show that silencing of STX8, particularly in the presence of the Vertex corrector molecule C18, rescues ΔF508-CFTR, allowing it to reach the cell surface and increasing CFTR-dependent chloride currents by approximately 2.5-fold over control values. STX8 silencing reduced the binding of quality control protein, Hsp 27, a protein that targets ΔF508-CFTR for sumoylation and subsequent degradation, to ΔF508-CFTR. STX8 silencing increased the levels of Hsp 60 a protein involving in early events in protein folding. CONCLUSION: STX8 knockdown creates an environment favorable for mature ΔF508 to reach the cell surface. The data also suggest that when present at normal levels, STX8 functions as part of the cell's quality control mechanism.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/metabolism , Endoplasmic Reticulum/metabolism , Qa-SNARE Proteins/metabolism , Cell Line , Cystic Fibrosis/genetics , Cystic Fibrosis/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/analysis , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/pathology , Gene Silencing , Humans , Protein Transport , Proteolysis , Qa-SNARE Proteins/analysis , Qa-SNARE Proteins/genetics
4.
J Cyst Fibros ; 17(5): 582-594, 2018 09.
Article in English | MEDLINE | ID: mdl-29936070

ABSTRACT

The missing phenylalanine at position 508, located in nucleotide-binding domain (NBD1) of the cystic fibrosis transmembrane regulator (CFTR), is the most common cystic fibrosis mutation. Severe disease-causing mutations also occur in NBD2. To provide information on potential therapeutic strategies for mutations in NBD2, we used a combination of biochemical, cell biological and electrophysiological approaches and newly created cell lines to study two disease-causing NBD2 mutants, N1303K and S1235R. We observed that neither was sensitive to E64, a cysteine protease inhibitor. However, further investigation showed that when treated with a combination of correctors, C4 + C18, both mutants also responded to E64. Further exploration to assess aggresome throughput using the autophagy regulator LC3 as a marker showed that, in the absence of correctors, N1303K showed a stalled throughput of LC3-II to the aggresome. The throughput became active again after treatment with the corrector combination C4 + C18. Confocal microscopic studies showed that the N1303K and S1235R mutant proteins both co-localized with LC3, but this co-localization was abolished by the corrector combination and, to a lesser extent, by VX-809. Both the corrector combination and VX-809 increased the CFTR chloride channel function of both mutants. We conclude that correctors have a dual effect, particularly on N1303K: they improve trafficking and function at the plasma membrane and reduce the association with autophagosomes. After treatment with correctors persistent degradation by the autophagosome may limit restoration of function. Thus, mutations in NBD2 of CFTR, in contrast to ΔF508-CFTR, may require additional personalized strategies to rescue them.


Subject(s)
Aminopyridines/pharmacology , Autophagosomes/physiology , Benzodioxoles/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Leucine/analogs & derivatives , Animals , Autophagy , Biological Transport , Blotting, Western , Cell Line , Electric Conductivity , Leucine/pharmacology , Mutant Proteins/drug effects , Mutant Proteins/genetics , Mutation , Small Molecule Libraries/pharmacology
5.
Cell Physiol Biochem ; 45(2): 639-655, 2018.
Article in English | MEDLINE | ID: mdl-29402832

ABSTRACT

BACKGROUND/AIMS: The CFTR-Associated Ligand (CAL), a PDZ domain containing protein with two coiled-coil domains, reduces cell surface WT CFTR through degradation in the lysosome by a well-characterized mechanism. However, CAL's regulatory effect on ΔF508 CFTR has remained almost entirely uninvestigated. METHODS: In this study, we describe a previously unknown pathway for CAL by which it regulates the membrane expression of ΔF508 CFTR through arrest of ΔF508 CFTR trafficking in the endoplasmic reticulum (ER) using a combination of cell biology, biochemistry and electrophysiology. RESULTS: We demonstrate that CAL is an ER localized protein that binds to ΔF508 CFTR and is degraded in the 26S proteasome. When CAL is inhibited, ΔF508 CFTR retention in the ER decreases and cell surface expression of mature functional ΔF508 CFTR is observed alongside of enhanced expression of plasma membrane scaffolding protein NHERF1. Chaperone proteins regulate this novel process, and ΔF508 CFTR binding to HSP40, HSP90, HSP70, VCP, and Aha1 changes to improve ΔF508 CFTR cell surface trafficking. CONCLUSION: Our results reveal a pathway in which CAL regulates the cell surface availability and intracellular retention of ΔF508 CFTR.


Subject(s)
Carrier Proteins/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/pathology , Membrane Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , COS Cells , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Line , Cell Membrane/metabolism , Chlorocebus aethiops , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Endoplasmic Reticulum/metabolism , Golgi Matrix Proteins , HSP40 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Humans , Macrolides/pharmacology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Transport Proteins , Phosphoproteins/metabolism , Protein Binding , Protein Transport/drug effects , RNA Interference , RNA, Ribosomal/metabolism , RNA, Small Interfering/metabolism , Sodium-Hydrogen Exchangers/metabolism
6.
Cell Physiol Biochem ; 41(6): 2194-2210, 2017.
Article in English | MEDLINE | ID: mdl-28448979

ABSTRACT

BACKGROUND/AIMS: Premature degradation of mutated cystic fibrosis transmembrane conductance regulator (CFTR) protein causes cystic fibrosis (CF), the commonest Mendelian disease in Caucasians. Despite recent advances in precision medicines for CF patients, many CFTR mutants have not been characterized and the effects of these new therapeutic approaches are still unclear for those mutants. METHODS: Cells transfected or stably expressing four CFTR transmembrane-domain mutants (G85E, E92K, L1077P, and M1101K) were used to: 1) characterize the mutants according to their protein expression, thermal sensitivity, and degradation pathways; 2) evaluate the effects of correctors in rescuing them; and 3) explore the effects of correctors on CFTR interactions with proteostasis components. RESULTS: All four mutants exhibited lower protein expression than did wild type-CFTR, and they were degraded by proteasomes and aggresomes. At low temperature, only cells expressing the mutants L1077P and M1101K exhibited increased CFTR maturation. Co-administration of C4 and C18 showed the greatest effect, restoring functional expression and partial stability of CFTR bearing E92K, L1077P, or M1101K at the cell surface. However, this treatment was inefficient in rectifying the defect of CFTR bearing G85E. Correctors rescued CFTR mutants by reducing their interactions with proteostasis components associated with protein retention in the endoplasmic reticulum and ubiquitination. CONCLUSION: Co-administration of C4 and C18 rescued CFTR transmembrane-domain mutants by remodeling the CFTR interactome.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Proteasome Endopeptidase Complex/metabolism , Anilides/pharmacology , Animals , COS Cells , Chlorocebus aethiops , Cysteine Proteinase Inhibitors/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Endoplasmic Reticulum/metabolism , HEK293 Cells , Humans , Hydroxamic Acids/pharmacology , Immunoprecipitation , Leupeptins/pharmacology , Mutagenesis, Site-Directed , Proteasome Endopeptidase Complex/chemistry , Protein Binding , Protein Stability , Proteolysis/drug effects , Temperature , Transfection
7.
Chembiochem ; 17(6): 493-505, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26864378

ABSTRACT

We evaluated whether small molecule correctors could rescue four nucleotide-binding domain 1 (NBD1) mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene (A455E, S492F, ΔI507, and R560T). We first transfected Cos-7 cells (green monkey kidney cells) with A455E, S492F, ΔI507, or R560T and created HEK-293 (human embryonic kidney cells) cell lines stably expressing these CFTR mutations. The mutants showed lowered protein expression, instability at physiological temperature, and rapid degradation. After treatment with correctors CFFT-002, CFFT-003, C3, C4, and/or C18, the combination of C18+C4 showed the most correction and resulted in increased CFTR residing in the plasma membrane. We found a profound decrease in binding of CFTR to histone deacetylases (HDAC) 6 and 7 and heat shock proteins (Hsps) 27 and 40. Silencing Hsp27 or 40 rescued the mutants, but no additional amount of CFTR was rescued when both proteins were knocked down simultaneously. Thus, CFTR mutations in NBD1 can be rescued by a combination of correctors, and the treatment alters the interaction between mutated CFTR and the endoplasmic reticulum machinery.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Mutation , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , HEK293 Cells , Humans
8.
J Biol Chem ; 290(42): 25636-45, 2015 Oct 16.
Article in English | MEDLINE | ID: mdl-26336106

ABSTRACT

Correcting the processing of ΔF508-CFTR, the most common mutation in cystic fibrosis, is the major goal in the development of new therapies for this disease. Here, we determined whether ΔF508 could be rescued by a combination of small-molecule correctors, and identified the mechanism by which correctors rescue the trafficking mutant of cystic fibrosis transmembrane conductance regulator (CFTR). We transfected COS-7 cells with ΔF508, created HEK-293 stably expressing ΔF508, and utilized CFBE41o(-) cell lines stably transduced with ΔF508. As shown previously, ΔF508 expressed less protein, was unstable at physiological temperature, and rapidly degraded. When the cells were treated with the combination C18 + C4 the mature C-band was expressed at the cell surface. After treatment with C18 + C4, we saw a lower rate of protein disappearance after translation was stopped with cycloheximide. To understand how this rescue occurs, we evaluated the change in the binding of proteins involved in endoplasmic reticulum-associated degradation, such as Hsp27 (HspB1) and Hsp40 (DnaJ). We saw a dramatic reduction in binding to heat shock proteins 27 and 40 following combined corrector therapy. siRNA experiments confirmed that a reduction in Hsp27 or Hsp40 rescued CFTR in the ΔF508 mutant, but the rescue was not additive or synergistic with C4 + 18 treatment, indicating these correctors shared a common pathway for rescue involving a network of endoplasmic reticulum-associated degradation proteins.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , HSP27 Heat-Shock Proteins/metabolism , HSP40 Heat-Shock Proteins/metabolism , Animals , COS Cells , Chlorocebus aethiops , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , HEK293 Cells , Humans , Mutation , Protein Binding , Temperature
9.
J Biol Chem ; 290(32): 19743-55, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26092729

ABSTRACT

Stargardt disease is the most common form of early onset macular degeneration. Mutations in ABCA4, a member of the ATP-binding cassette (ABC) family, are associated with Stargardt disease. Here, we have examined two disease-causing mutations in the NBD1 region of ABCA4, R1108C, and R1129C, which occur within regions of high similarity with CFTR, another ABC transporter gene, which is associated with cystic fibrosis. We show that R1108C and R1129C are both temperature-sensitive processing mutants that engage the cellular quality control mechanism and show a strong interaction with the chaperone Hsp 27. Both mutant proteins also interact with HDCAC6 and are degraded in the aggresome. We also demonstrate that novel corrector compounds that are being tested as treatment for cystic fibrosis, such as VX-809, can rescue the processing of the ABCA4 mutants, particularly their expression at the cell surface, and can reduce their binding to HDAC6. Thus, our data suggest that VX-809 can potentially be developed as a new therapy for Stargardt disease, for which there is currently no treatment.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Aminopyridines/pharmacology , Benzodioxoles/pharmacology , HSP27 Heat-Shock Proteins/metabolism , Histone Deacetylases/metabolism , Protective Agents/pharmacology , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/genetics , Amino Acid Sequence , Anilides/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression , HEK293 Cells , HSP27 Heat-Shock Proteins/genetics , Histone Deacetylase 6 , Histone Deacetylases/genetics , Humans , Hydroxamic Acids/pharmacology , Macrolides/pharmacology , Macular Degeneration/congenital , Macular Degeneration/drug therapy , Macular Degeneration/genetics , Macular Degeneration/metabolism , Molecular Sequence Data , Mutation , Protein Transport , Proteolysis , Sequence Homology, Amino Acid , Signal Transduction , Stargardt Disease , Transgenes
10.
PLoS One ; 10(3): e0119796, 2015.
Article in English | MEDLINE | ID: mdl-25799511

ABSTRACT

Although, the most common Cystic Fibrosis mutation, ΔF508, in the cystic fibrosis transmembrane regulator. (CFTR), is located in nucleotide binding domain (NBD1), disease-causing mutations also occur in NBD2. To provide information on potential therapeutic strategies for mutations in NBD2, we studied, using a combination of biochemical approaches and newly created cell lines, two disease-causing NBD2 mutants, N1303K and S1235R. Surprisingly, neither was rescued by low temperature. Inhibition of proteasomes with MG132 or aggresomes with tubacin rescued the immature B and mature C bands of N1303K and S1235R, indicating that degradation occurs via proteasomes and aggresomes. We found no effect of the lysosome inhibitor E64. Thus, our results show that these NBD2 mutants are processing mutants with unique characteristics. Several known correctors developed to rescue ΔF508-CFTR, when applied either alone or in combination, significantly increased the maturation of bands B and C of both NBD 2 mutants. The best correction occurred with the combinations of C4 plus C18 or C3 plus C4. Co-transfection of truncated CFTR (∆27-264) into stably transfected cells was also able to rescue them. This demonstrates for the first time that transcomplementation with a truncated version of CFTR can rescue NBD2 mutants. Our results show that the N1303K mutation has a more profound effect on NBD2 processing than S1235R and that small-molecule correctors increase the maturation of bands B and C in NBD2 mutants. In addition, ∆27-264 was able to transcomplement both NDB2 mutants. We conclude that differences and similarities occur in the impact of mutations on NBD2 when compared to ΔF508-CFTR suggesting that individualized strategies may be needed to restore their function. Finally our results are important because they suggest that gene or corrector molecule therapies either alone or in combination individualized for NBD2 mutants may be beneficial for patients bearing N1303K or S1235R mutations.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Genetic Complementation Test , Mutation/genetics , Small Molecule Libraries/pharmacology , Animals , Biological Transport , COS Cells , Chlorocebus aethiops , Cystic Fibrosis/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , HEK293 Cells , Humans , Protein Binding
11.
J Neurotrauma ; 30(16): 1449-56, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23611588

ABSTRACT

Brains undergo significant remodeling after traumatic brain injury (TBI). The Rho guanine triphosphate (GTP)ase pathways control brain remodeling during development and under pathological conditions. How the Rho GTPase pathways are regulated in the brain after TBI remains largely unknown, however. This study used the rat fluid percussion injury model to investigate changes in the Rho GTPase pathways after TBI. The results showed that TBI leads to activation and translocation of RhoA and Rac1 proteins from cytosolic fraction to the membrane fraction after injury. Consistently, the Rho guanine nucleotide exchange factors GEF-H1 and Cool-2/αPix are significantly activated by dephosphorylation and accumulation in the cytosolic fractions during the post-TBI phase. Because the Rho GTPase pathways are key regulators of brain remodeling, these results depict regulatory mechanisms of the Rho GTPase pathways after TBI, and pave the way for the study of therapeutic agents targeting the Rho GTPase pathways for functional recovery after TBI.


Subject(s)
Brain Injuries/metabolism , Brain Injuries/pathology , Rho Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction/physiology , Animals , Brain/metabolism , Brain/pathology , Male , Rats , Rats, Sprague-Dawley , rho GTP-Binding Proteins/metabolism
12.
J Mol Neurosci ; 44(1): 68-75, 2011 May.
Article in English | MEDLINE | ID: mdl-21437668

ABSTRACT

RNA interference (RNAi) is the process of sequence-specific posttranslational gene silencing triggered by double-stranded RNAs (dsRNAs). RNAi is a widely used approach for studying gene function. However, studies have shown that using siRNA can lead to off-target effects when the siRNA contains sufficient sequence identity to non-target mRNA sequences. One of the important steps in designing dsRNA is verification that it has sequence identity to only the target mRNA. In this report, we propose an approach for primary screening dsRNAs for potential off-target effects by using rapid amplification of cDNA ends. This method can be especially useful for model systems using species that have limited availability of sequence data.


Subject(s)
Nucleic Acid Amplification Techniques , RNA Interference , RNA, Small Interfering/genetics , Animals , DNA, Complementary/genetics , RNA, Double-Stranded/genetics , Turtles
13.
Biochem Biophys Res Commun ; 389(2): 338-42, 2009 Nov 13.
Article in English | MEDLINE | ID: mdl-19723501

ABSTRACT

Mammalian Tolloid-like 1 (mTll-1) is an astacin metalloprotease that is a member of the Tolloid family of proteins. mTll-1 cleaves chordin, an inhibitor of bone morphogenetic proteins (BMPs) and potentiates activity of the BMPs. Prenatal stress and glucocorticoids decrease mTll-1 expression whereas voluntary exercise increase mTll-1 gene expression in the mouse hippocampus. Here, we studied the underlying molecular mechanisms by which hypoxia regulates human mTll-1 gene expression. When cells were subjected to hypoxia, the expression of endogenous mTll-1 was upregulated in SH-SY5Y human neuroblastoma cells. Dual-luciferase assay and site-directed mutagenesis showed the presence of hypoxia responsive elements (HREs) at position 625 that was essential for activation of mTll-1 expression under hypoxic conditions. The binding of hypoxia-inducible factor (HIF-1) protein to the HREs was confirmed by gel shift assay. These results indicate that the HRE motif is directly involved in the activation of the mTll-1 transcription under hypoxic conditions.


Subject(s)
Gene Expression Regulation , Hypoxia-Inducible Factor 1/metabolism , Tolloid-Like Metalloproteinases/genetics , Animals , Binding Sites , Cell Hypoxia/genetics , Cell Line, Tumor , Humans , Mice , Regulatory Elements, Transcriptional , Up-Regulation
14.
Dev Growth Differ ; 51(4): 403-10, 2009 May.
Article in English | MEDLINE | ID: mdl-19366374

ABSTRACT

Mammalian Tolloid-like 1 (Tll-1) is a pleiotropic metalloprotease that is expressed by a small subset of cells within the precardiac mesoderm and is necessary for proper heart development. Following heart tube formation Tll-1 is expressed by the endocardium and regions of myocardium overlying the region of the muscular interventricular septum. Mutations in Tll-1 lead to embryonic lethality due to cardiac defects. We demonstrate that the Tll-1promoter contains Nkx2-5 binding sites and that the Tll-1 promoter is activated by and directly binds Nkx2-5.Tll-1 expression is ablated by a dominant negative Nkx2-5 or by mutation of the Nkx2-5 binding sites within theTll-1 promoter. In vivo, Tll-1 expression is decreased in the hearts of Nkx2-5 knockout embryos when compared with hemizygous and wild-type embryos. These results show that Nkx2-5 is a direct activator of Tll-1 expression and provide insight into the mechanism of the defects found in both the Tll-1 and Nkx2-5 knockout mice.


Subject(s)
Homeodomain Proteins/metabolism , Myocardium/metabolism , Tolloid-Like Metalloproteinases/metabolism , Transcription Factors/metabolism , Animals , Cell Line , Electrophoretic Mobility Shift Assay , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Mice , Polymerase Chain Reaction , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Quail , Tolloid-Like Metalloproteinases/genetics , Transcription Factors/genetics
15.
Plant Physiol Biochem ; 43(4): 419-22, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15907695

ABSTRACT

Adding PEG to the nutrient medium of maize (Zea mays L., hybrid Harkovskaya 310 MV) plants arrested the growth of their leaves initially but in 40-50 min growth resumed. This coincided with and was obviously due to a gradual increase in extensibility of the primary leaf suggested by changes in its extension rate, which was induced by adding a counterweight to inductive electromechanical position sensor. Specificity of gene probe for expansins was confirmed by sequencing cDNA and its comparison with literature data. Dot-blot analysis showed an increase in transcript level of expansin genes induced by PEG treatment. Thus gene-specific regulation of expansin mRNA pools likely contributes to fast adjustment of cell wall-loosening under conditions of water deficit.


Subject(s)
Plant Proteins/genetics , Zea mays/genetics , Zea mays/metabolism , Base Sequence , DNA, Plant/genetics , Gene Expression/drug effects , Genes, Plant , Osmotic Pressure , Plant Leaves/drug effects , Plant Leaves/growth & development , Plant Leaves/metabolism , Polyethylene Glycols/pharmacology , Zea mays/drug effects , Zea mays/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...