Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Biochemistry ; 62(17): 2597-2610, 2023 09 05.
Article in English | MEDLINE | ID: mdl-37556352

ABSTRACT

Purine-rich element binding protein B (Purß) is a single-stranded DNA (ssDNA) and RNA-binding protein that functions as a transcriptional repressor of genes encoding certain muscle-restricted contractile proteins in the setting of cellular stress or tissue injury. A prior report from our laboratory implicated specific basic amino acid residues in the physical and functional interaction of Purß with the smooth muscle-α actin gene (Acta2) promoter. Independent structural analysis of fruit fly Purα uncovered a role for several aromatic residues in the binding of this related protein to ssDNA. Herein, we examine the functional importance of a comparable set of hydrophobic residues that are positionally conserved in the repeat I (Y59), II (F155), and III (F256) domains of murine Purß. Site-directed Y/F to alanine substitutions were engineered, and the resultant Purß point mutants were tested in various biochemical and cell-based assays. None of the mutations affected the cellular expression, structural stability, or dimerization capacity of Purß. However, the Y59A and F155A mutants demonstrated weaker Acta2 repressor activity in transfected fibroblasts and reduced binding affinity for the purine-rich strand of an Acta2 cis-regulatory element in vitro. Mutation of Y59 and F155 also altered the multisite binding properties of Purß for ssDNA and diminished the interaction of Purß with Y-box binding protein 1, a co-repressor of Acta2. Collectively, these findings suggest that some of the same aromatic residues, which govern the specific and high-affinity binding of Purß to ssDNA, also mediate certain heterotypic protein interactions underlying the Acta2 repressor function of Purß.


Subject(s)
DNA, Single-Stranded , DNA-Binding Proteins , Mice , Animals , DNA-Binding Proteins/chemistry , DNA, Single-Stranded/genetics , Promoter Regions, Genetic , Purines , Protein Binding
2.
Mol Cell Biochem ; 476(8): 2899-2916, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33743134

ABSTRACT

Myocardin is a potent transcriptional coactivator protein, which functions as the master regulator of vascular smooth muscle cell differentiation. The cofactor activity of myocardin is mediated by its physical interaction with serum response factor, a ubiquitously expressed transactivator that binds to CArG boxes in genes encoding smooth muscle-restricted proteins. Purine-rich element binding protein B (Purß) represses the transcription of the smooth muscle α-actin gene (Acta2) in fibroblasts and smooth muscle cells by interacting with single-stranded DNA sequences flanking two 5' CArG boxes in the Acta2 promoter. In this study, the ability of Purß to modulate the cofactor activity of myocardin was investigated using a combination of cellular and biochemical approaches. Results of smooth muscle gene promoter-reporter assays indicated that Purß specifically inhibits the coactivator function of myocardin in a manner requiring the presence of all three single-stranded DNA binding domains in the Purß homodimer. DNA binding analyses demonstrated that Purß interacts with CArG-containing DNA elements with a much lower affinity compared to other purine-rich target sequences present in the Acta2 promoter. Co-immunoprecipitation and DNA pull-down assays revealed that Purß associates with myocardin and serum response factor when free or bound to duplex DNA containing one or more CArG boxes. Functional analysis of engineered Purß point mutants identified several amino acid residues essential for suppression of myocardin activity. Collectively, these findings suggest an inhibitory mechanism involving direct protein-protein interaction between the homodimeric Purß repressor and the myocardin-serum response factor-CArG complex.


Subject(s)
Cell Differentiation , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Muscle Proteins/metabolism , Muscle, Smooth/cytology , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Actins/genetics , Actins/metabolism , Animals , DNA-Binding Proteins/genetics , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Muscle Proteins/genetics , Muscle, Smooth/metabolism , Nuclear Proteins/genetics , Promoter Regions, Genetic , Protein Binding , Purines/metabolism , Rats , Trans-Activators/genetics
3.
J Cell Biochem ; 120(4): 5835-5851, 2019 04.
Article in English | MEDLINE | ID: mdl-30387171

ABSTRACT

Purine-rich element-binding protein B (Purß) inhibits myofibroblast differentiation by repressing the expression of the smooth muscle α-actin gene (Acta2). Several reports have identified the structural domains in Purß that enable its characteristic interaction with purine-rich single-stranded DNA (ssDNA) sequences in the Acta2 promoter. However, little is known about the physical and functional effects of single-nucleotide polymorphisms that alter individual amino acid residues in Purß. This study evaluated seven rare single amino acid variants of human PURB engineered into the homologous mouse Purß protein. Mapping the location of variant residues on a homology model of the Purß homodimer suggested that most of the altered residues are remote from the predicted ssDNA-binding regions of the protein. The repressor activity of each Purß variant was assessed in transfected fibroblasts and smooth muscle cells via Acta2 promoter-reporter assays. A Q64* nonsense variant was completely inactive while missense variants exhibited repressor activity that ranged from ~1.5-fold greater to ~2-fold less than wild-type Purß. Lower activity variants P223L and R297Q were expressed in bacteria and purified to homogeneity. Each variant was physically indistinguishable from wild-type Purß in terms of quaternary structure and thermostability. Results of DNA and protein-binding assays indicated that the P223L and R297Q variants retained high affinity and specificity for purine-rich ssDNA sequences but differed in their interaction with other Acta2 regulatory proteins. These findings suggest that the presence of certain variant residues affects the Acta2 repressor activity of Purß by altering its interaction with other transcription factors but not with ssDNA.


Subject(s)
Actins/metabolism , Codon, Nonsense , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Mutant Proteins/metabolism , Polymorphism, Single Nucleotide , Amino Acid Sequence , Animals , DNA-Binding Proteins/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Mice , Mutant Proteins/chemistry , Mutant Proteins/genetics , Promoter Regions, Genetic , Protein Binding , Protein Conformation , Sequence Homology
4.
J Cell Biochem ; 119(2): 2073-2083, 2018 02.
Article in English | MEDLINE | ID: mdl-28834593

ABSTRACT

Acute myelogenous leukemia (AML) is an aggressive hematologic cancer characterized by infiltration of proliferative, clonal, abnormally differentiated cells of myeloid lineage in the bone marrow and blood. Malignant cells in AML often exhibit chromosomal and other genetic or epigenetic abnormalities that are useful in prognostic risk assessment. In this study, the relative expression and novel single-stranded DNA (ssDNA) binding function of purine-rich element binding proteins A and B (Purα and Purß) were systematically evaluated in established leukemia cell lines and in lineage committed myeloid cells isolated from patients diagnosed with a hematologic malignancy. Western blotting revealed that Purα and Purß are markedly elevated in CD33+ /CD66b+ cells from AML patients compared to healthy subjects and to patients with other types of myeloid cell disorders. Results of in silico database analysis of PURA and PURB mRNA expression during hematopoiesis in conjunction with the quantitative immunoassay of the ssDNA-binding activities of Purα and Purß in transformed leukocyte cell lines pointed to Purß as the more distinguishing biomarker of myeloid cell differentiation status. Purß ssDNA-binding activity was significantly increased in myeloid cells from AML patients but not from individuals with other myeloid-related diseases. The highest levels of Purß activity were detected in myeloid cells from primary AML patients and from AML patients displaying other risk factors forecasting a poor prognosis. Collectively, these findings suggest that the enhanced ssDNA-binding activity of Purß in transformed myeloid cells may serve as a unique and measurable phenotypic trait for improving prognostic risk stratification in AML.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , DNA, Single-Stranded/metabolism , Female , Gene Expression Regulation, Leukemic , Humans , Male , Middle Aged , Prognosis , Protein Binding , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Am J Hematol ; 92(11): 1119-1130, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28699284

ABSTRACT

Elaboration of tumor necrosis factor (TNF) is a very early event in development of ischemia/reperfusion injury pathophysiology. Therefore, TNF may be a prominent mediator of endothelial cell and vascular wall dysfunction in sickle cell anemia, a hypothesis we addressed using NY1DD, S+SAntilles , and SS-BERK sickle transgenic mice. Transfusion experiments revealed participation of abnormally activated blood monocytes exerting an endothelial activating effect, dependent upon Egr-1 in both vessel wall and blood cells, and upon NFκB(p50) in a blood cell only. Involvement of TNF was identified by beneficial impact from TNF blockers, etanercept and infliximab, with less benefit from an IL-1 blocker, anakinra. In therapeutic studies, etanercept ameliorated multiple disturbances of the murine sickle condition: monocyte activation, blood biomarkers of inflammation, low platelet count and Hb, vascular stasis triggered by hypoxia/reoxygenation (but not if triggered by hemin infusion), tissue production of neuro-inflammatory mediators, endothelial activation (monitored by tissue factor and VCAM-1 expression), histopathologic liver injury, and three surrogate markers of pulmonary hypertension (perivascular inflammatory aggregates, arteriolar muscularization, and right ventricular mean systolic pressure). In aggregate, these studies identify a prominent-and possibly dominant-role for an abnormal monocyte-TNF-endothelial activation axis in the sickle context. Its presence, plus the many benefits of etanercept observed here, argue that pilot testing of TNF blockade should be considered for human sickle cell anemia, a challenging but achievable translational research goal.


Subject(s)
Anemia, Sickle Cell/metabolism , Endothelial Cells/metabolism , Monocytes/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Anemia, Sickle Cell/diagnosis , Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/genetics , Animals , Antibodies, Monoclonal/pharmacology , Biomarkers , Bone Marrow Transplantation , Cell Aggregation/genetics , Cell Aggregation/immunology , Disease Models, Animal , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Endothelium, Vascular/metabolism , Etanercept/pharmacology , Etanercept/therapeutic use , Heart Function Tests , Humans , Inflammation Mediators , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Molecular Targeted Therapy , Monocytes/drug effects , Monocytes/immunology , NF-kappa B/deficiency , NF-kappa B/genetics , Phenotype , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Thromboplastin/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Vascular Cell Adhesion Molecule-1/metabolism
6.
Biochemistry ; 55(19): 2794-805, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27064749

ABSTRACT

Myofibroblast differentiation is characterized by an increased level of expression of cytoskeletal smooth muscle α-actin. In human and murine fibroblasts, the gene encoding smooth muscle α-actin (Acta2) is tightly regulated by a network of transcription factors that either activate or repress the 5' promoter-enhancer in response to environmental cues signaling tissue repair and remodeling. Purine-rich element-binding protein B (Purß) suppresses the expression of Acta2 by cooperatively interacting with the sense strand of a 5' polypurine sequence containing an inverted MCAT cis element required for gene activation. In this study, we evaluated the chemical basis of nucleoprotein complex formation between the Purß repressor and the purine-rich strand of the MCAT element in the mouse Acta2 promoter. Quantitative single-stranded DNA (ssDNA) binding assays conducted in the presence of increasing concentrations of monovalent salt or anionic detergent suggested that the assembly of a high-affinity nucleoprotein complex is driven by a combination of electrostatic and hydrophobic interactions. Consistent with the results of pH titration analysis, site-directed mutagenesis revealed several basic amino acid residues in the intermolecular (R267) and intramolecular (K82 and R159) subdomains that are essential for Purß transcriptional repressor function in Acta2 promoter-reporter assays. In keeping with their diminished Acta2 repressor activity in fibroblasts, purified Purß variants containing an R267A mutation exhibited reduced binding affinity for purine-rich ssDNA. Moreover, certain double and triple-point mutants were also defective in binding to the Acta2 corepressor protein, Y-box-binding protein 1. Collectively, these findings establish the repertoire of noncovalent interactions that account for the unique structural and functional properties of Purß.


Subject(s)
Actins , DNA, Single-Stranded , DNA-Binding Proteins , Fibroblasts/metabolism , Multiprotein Complexes , Actins/chemistry , Actins/genetics , Actins/metabolism , Animals , Cell Line , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fibroblasts/cytology , Humans , Mice , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Protein Domains , Protein Structure, Quaternary , Static Electricity
7.
J Cell Physiol ; 229(9): 1256-71, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24446247

ABSTRACT

Expression of smooth muscle alpha-actin (SMαA) is essential for myofibroblast-mediated wound contraction following tissue injury. The Pur α/ß and YB-1 transcriptional repressors govern the DNA-binding activity of serum response factor (SRF) and phosphorylated Smad3 (pSmad3) transcriptional activators during induction of SMαA gene expression in human pulmonary myofibroblasts. In quiescent fibroblasts, Pur α exhibited a novel function in enhancing stability of pre-existing SRF complexes with SMαA core promoter DNA, whereas Pur ß was more effective in disrupting SRF-DNA interaction. Pur proteins were less efficient competitors of pre-existing, core-promoter complexes containing both SRF and pSmad3 in nuclear extracts from TGFß1-activated myofibroblasts. TGFß1 signaling dissociated a SRF/Pur protein complex with concurrent formation of a transient pSmad3/MRTF-A/Pur ß complex during early phase myofibroblast differentiation. Pur ß was replaced by Pur α in the pSmad3/MRTF-A complex in mature myofibroblasts. Combining all three repressors potently inhibited SRF and pSmad3 binding to promoter DNA in quiescent fibroblasts and TGFß1-activated myofibroblasts, respectively. The results point to dynamic interplay between transcriptional activators and repressors in regulating SMαA gene output during myofibroblast differentiation. Therapeutic targeting of nucleoprotein complexes regulating the SMαA promoter may prevent excessive myofibroblast accumulation associated with chronic cardiopulmonary fibrosis and dysfunctional tissue remodeling.


Subject(s)
Actins/metabolism , Cell Differentiation , DNA-Binding Proteins/metabolism , Lung/metabolism , Myofibroblasts/metabolism , Pulmonary Fibrosis/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Actins/genetics , Binding Sites , Cells, Cultured , DNA-Binding Proteins/genetics , Fibrosis , Humans , Lung/pathology , Myofibroblasts/pathology , Oncogene Proteins, Fusion/metabolism , Phosphorylation , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , RNA Interference , Serum Response Element , Serum Response Factor/metabolism , Signal Transduction , Smad3 Protein/metabolism , Time Factors , Trans-Activators , Transcription Factors/genetics , Transfection , Transforming Growth Factor beta1/metabolism , Up-Regulation
8.
Biochemistry ; 52(26): 4439-50, 2013 Jul 02.
Article in English | MEDLINE | ID: mdl-23724822

ABSTRACT

A hallmark of dysfunctional fibroblast to myofibroblast differentiation associated with fibrotic disorders is persistent expression of ACTA2, the gene encoding the cyto-contractile protein smooth muscle α-actin. In this study, a PURB-specific gene knockdown approach was used in conjunction with biochemical analyses of protein subdomain structure and function to reveal the mechanism by which purine-rich element binding protein B (Purß) restricts ACTA2 expression in mouse embryo fibroblasts (MEFs). Consistent with the hypothesized role of Purß as a suppressor of myofibroblast differentiation, stable short hairpin RNA-mediated knockdown of Purß in cultured MEFs promoted changes in cell morphology, actin isoform expression, and cell migration indicative of conversion to a myofibroblast-like phenotype. Promoter-reporter assays in transfected Purß knockdown MEFs confirmed that these changes were attributable, in part, to derepression of ACTA2 transcription. To map the domains in Purß responsible for ACTA2 repression, several recombinant truncation mutants were generated and analyzed based on hypothetical, computationally derived models of the tertiary and quaternary structure of Purß. Discrete subdomains mediating sequence- and strand-specific cis-element binding, protein-protein interaction, and inhibition of a composite ACTA2 enhancer were identified using a combination of biochemical, biophysical, and cell-based assays. Our results indicate that the Purß homodimer possesses three separate but unequal single-stranded DNA-binding modules formed by subdomain-specific inter- and intramolecular interactions. This structural arrangement suggests that the cooperative assembly of the dimeric Purß repressor on the sense strand of the ACTA2 enhancer is dictated by the association of each subdomain with distinct purine-rich binding sites within the enhancer.


Subject(s)
Actins/chemistry , Cell Differentiation/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Actins/metabolism , Animals , Binding Sites , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/genetics , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Fibroblasts/metabolism , Mice , Myofibroblasts/metabolism , Promoter Regions, Genetic , Protein Binding , Purines/chemistry
9.
Exp Biol Med (Maywood) ; 237(5): 593-607, 2012 May.
Article in English | MEDLINE | ID: mdl-22619371

ABSTRACT

Peri-transplant surgical trauma and ischemia/reperfusion injury in accepted murine heterotopic heart grafts has been associated with myofibroblast differentiation, cardiac fibrosis and biomechanical-stress activation of the fetal myocardial smooth muscle α-actin (SMαA) gene. The wound-healing agonists, transforming growth factor ß1 and thrombin, are known to coordinate SMαA mRNA transcription and translation in activated myofibroblasts by altering the subcellular localization and mRNA-binding affinity of the Y-box binding protein-1 (YB-1) cold-shock domain (CSD) protein that governs a variety of cellular responses to metabolic stress. YB-1 accumulated in polyribosome-enriched regions of the sarcoplasm proximal to cardiac intercalated discs in accepted heart grafts. YB-1 binding to a purine-rich motif in exon 3 of SMαA mRNA that regulates translational efficiency increased substantially in perfusion-isolated, rod-shaped adult rat cardiomyocytes during phenotypic de-differentiation in the presence of serum-derived growth factors. Cardiomyocyte de-differentiation was accompanied by the loss of a 60 kDa YB-1 variant that was highly expressed in both adult myocardium and freshly isolated myocytes and replacement with the 50 kDa form of YB-1 (p50) typically expressed in myofibroblasts that demonstrated sequence-specific interaction with SMαA mRNA. Accumulation of p50 YB-1 in reprogrammed, de-differentiated myocytes was associated with a 10-fold increase in SMαA protein expression. Endomyocardial biopsies collected from patients up to 14 years after heart transplant showed variable yet coordinately elevated expression of SMαA and p50 YB-1 protein and demonstrable p50 YB-1:SMαA mRNA interaction. The p60 YB-1 variant in human heart graft samples, but neither mouse p60 nor mouse or human p50, reacted with an antibody specific for the phosphoserine 102 modification in the YB-1 CSD. Modulation of YB-1 subcellular compartmentalization and mRNA-binding activity may be linked with reprogramming of contractile protein gene expression in ventricular cardiomyocytes that could contribute to maladaptive remodeling in accepted, long-term heart grafts.


Subject(s)
Heart Transplantation , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Y-Box-Binding Protein 1/metabolism , Actins/genetics , Actins/metabolism , Animals , Animals, Newborn , Electrophoretic Mobility Shift Assay , Gene Expression , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Muscle, Smooth, Vascular/metabolism , Myocardial Reperfusion Injury/metabolism , Myofibroblasts/metabolism , Promoter Regions, Genetic , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Transcription, Genetic , Transplantation, Heterotopic , Wound Healing , Y-Box-Binding Protein 1/genetics
10.
Circulation ; 125(22): 2751-61, 2012 Jun 05.
Article in English | MEDLINE | ID: mdl-22570371

ABSTRACT

BACKGROUND: Delineating the role of microRNAs (miRNAs) in the posttranscriptional gene regulation offers new insights into how the heart adapts to pathological stress. We developed a knockout of miR-22 in mice and investigated its function in the heart. METHODS AND RESULTS: Here, we show that miR-22-deficient mice are impaired in inotropic and lusitropic response to acute stress by dobutamine. Furthermore, the absence of miR-22 sensitized mice to cardiac decompensation and left ventricular dilation after long-term stimulation by pressure overload. Calcium transient analysis revealed reduced sarcoplasmic reticulum Ca(2+) load in association with repressed sarcoplasmic reticulum Ca(2+) ATPase activity in mutant myocytes. Genetic ablation of miR-22 also led to a decrease in cardiac expression levels for Serca2a and muscle-restricted genes encoding proteins in the vicinity of the cardiac Z disk/titin cytoskeleton. These phenotypes were attributed in part to inappropriate repression of serum response factor activity in stressed hearts. Global analysis revealed increased expression of the transcriptional/translational repressor purine-rich element binding protein B, a highly conserved miR-22 target implicated in the negative control of muscle expression. CONCLUSION: These data indicate that miR-22 functions as an integrator of Ca(2+) homeostasis and myofibrillar protein content during stress in the heart and shed light on the mechanisms that enhance propensity toward heart failure.


Subject(s)
Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/physiopathology , MicroRNAs/genetics , MicroRNAs/metabolism , Myocardial Contraction/physiology , Stress, Physiological/physiology , Animals , Calcium/metabolism , Cardiomyopathy, Dilated/pathology , DNA-Binding Proteins/metabolism , Disease Models, Animal , Gene Expression Regulation/physiology , Homeostasis/physiology , Male , Mice , Mice, Knockout , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Serum Response Factor/metabolism
11.
Biochem Biophys Res Commun ; 400(3): 340-5, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20728429

ABSTRACT

Purß is a single-stranded nucleic acid-binding protein implicated in the injury-induced repression of genes encoding certain muscle-restricted isoforms of actin and myosin expressed in the heart, skeletal muscle, and vasculature. To better understand how the modular arrangement of the primary sequence of Purß affects the higher order structure and function of the protein, purified recombinant Purß was subjected to partial proteolysis in an attempt to identify a well-folded truncation protein that retained purine-rich single-stranded DNA-binding activity. Limited tryptic digestion of Purß liberated a core ∼30kDa fragment corresponding to residues 29-305 as determined by epitope mapping and mass spectrometry. Size exclusion chromatography indicated that the isolated core fragment retains the ability to self-associate while circular dichroism analysis confirmed that the Purß core domain is stably folded in the absence of glycine-rich N- and C-terminal sequences. Comparative DNA-binding assays revealed that the isolated core domain interacts with purine-rich cis-elements from the smooth muscle α-actin gene with similar specificity but increased affinity compared to full-length Purß. These findings suggest that the highly conserved modular repeats of Purß fold to form a core functional domain, which mediates the specific and high affinity binding of the protein to single-stranded DNA.


Subject(s)
DNA-Binding Proteins/chemistry , Animals , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/isolation & purification , Mice , Protein Folding , Protein Structure, Quaternary , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Trypsin/chemistry
12.
Exp Biol Med (Maywood) ; 235(8): 966-73, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20660097

ABSTRACT

We previously observed gross hemorrhage in plasminogen activator inhibitor type-1 (PAI-1) knockout (PKO) mice with induced myocardial infarction (MI). We hypothesized that it reflected degradation of vessels - a phenomenon we termed vascular rhexis. Accordingly, in the present study we characterized vascular rhexis in C57BL6 mice. MI was induced in 10- to 12-week-old mice by coronary artery ligation for 24, 48, 72 or 96 h. Hemorrhage was quantified by non-cross-reacting enzyme-linked immunosorbent assay of left ventricular (LV) hemoglobin corrected for myoglobin. Degradation of vasculature was quantified by the appearance of alpha smooth muscle actin (alphaSMA) in low salt soluble fractions of LV homogenates (Western blotting) and by immunohistochemistry (residual alphaSMA). Co-staining for CD31 (endothelial cells) and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling (TUNEL) (a marker of cell death) was used to identify capillary rhexis. PKO mice (n = 9) had marked hemorrhage in infarct zones (432 +/- 27 standard error of mean microL blood/g). Hemorrhage was evident in C57BL6 mice as well (n = 6): 51 +/- 8 microL/g LV 96 h after coronary occlusion compared with 10 +/- 5 microL /g, n = 13 in normal LVs. Residual intact vasculature was reduced 48 h after infarction. Thus, an average of 16 +/- 1.6 small- and medium-sized vessels (n = 5 hearts) were seen compared with 84 +/- 4.8 in normal LVs (n = 3, P < or = 0.05). An approximately three-fold increase in soluble alphaSMA 48 h after MI (2.68 +/- 0.28, n = 6) was seen relative to that in normal LVs defined as 1.0 +/- 0.04, n = 10, P < or = 0.05. Capillary degradation was evident as well, as judged from CD31 and TUNEL co-localization. Vascular rhexis occurs within 48 h after the onset of MI. It may contribute to the early no-reflow phenomenon and to late negative LV remodeling.


Subject(s)
Coronary Vessels/pathology , Coronary Vessels/physiopathology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Animals , Blotting, Western , Coronary Vessels/metabolism , Enzyme-Linked Immunosorbent Assay , Heart Failure/metabolism , Heart Failure/physiopathology , Hemorrhage/metabolism , Immunohistochemistry , Ischemia/metabolism , Ischemia/physiopathology , Mice , Mice, Inbred C57BL , Microcirculation/physiology , Myocardial Infarction/metabolism , Neovascularization, Physiologic , Plasminogen Activator Inhibitor 1/metabolism , Time Factors , Ventricular Dysfunction, Left , Ventricular Remodeling
13.
Transl Res ; 155(4): 170-7, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20303465

ABSTRACT

Sickle cell anemia is accompanied by the activation of coagulation and thrombosis. We have studied the abnormal expression of tissue factor (TF) by the pulmonary vein endothelium of the mild-phenotype NY1DD sickle transgenic. As detected by immunofluorescence microscopy, this occurs only after the NY1DD mouse is exposed to hypoxia/reoxygenation (H/R), which actually causes ischemia/reperfusion in the sickle cell disease-but not the normal-mouse model. We tested the hypothesis that the nuclear factor-kappa B (NFkappaB)-activating inflammation that develops in post-H/R NY1DD mice is responsible for this phenotype switch. Various NFkappaB inhibitors (including p50-specific andrographolide) demonstrated that endothelial TF positivity is NFkappaB dependent. Several systemic inflammatory stimulators (tumor necrosis factor [TNFalpha], lipopolysaccharide, thioglycollate, and carageenan) given to control mice showed that the inflammatory promotion of TF expression by only pulmonary vein endothelium is not specific to the sickle cell disease model. We bred the NFkappaB(p50)-/- state into the NY1DD mouse. Combined with marrow transplantation, this allowed the creation of NY1DD mice that were NFkappaB(p50)-/- only in peripheral blood cells (and marrow) versus only in vessel walls (and tissues). This process revealed that endothelial TF expression in the NY1DD mouse is highly dependent on NFkappaB(p50) in peripheral blood mononuclear cells-but not in the vessel wall. In confirmation, the infusion of post-H/R sickle mouse blood mononuclear cells into naïve NY1DD mice stimulated endothelial TF expression; the infusion of such cells from unstimulated sickle cell disease mice at ambient air did not stimulate TF expression. We conclude that peripheral blood mononuclear cells indirectly promote endothelial TF expression via a NFkappaB(p50)-dependent mechanism. This approach may be relevant to the role of coagulopathy in clinical sickle cell disease.


Subject(s)
Anemia, Sickle Cell/blood , Blood Coagulation/physiology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Leukocytes, Mononuclear/metabolism , NF-kappa B p50 Subunit/metabolism , Thromboplastin/metabolism , Anemia, Sickle Cell/complications , Animals , Gene Knockout Techniques , Humans , Inflammation/metabolism , Inflammation/pathology , Leukocytes, Mononuclear/pathology , Mice , Mice, Transgenic , NF-kappa B p50 Subunit/antagonists & inhibitors , NF-kappa B p50 Subunit/deficiency
14.
Blood ; 115(12): 2483-90, 2010 Mar 25.
Article in English | MEDLINE | ID: mdl-20053759

ABSTRACT

The vascular pathobiology of sickle cell anemia involves inflammation, coagulation, vascular stasis, reperfusion injury, iron-based oxidative biochemistry, deficient nitric oxide (NO) bioavailability, and red cell sickling. These disparate pathobiologies intersect and overlap, so it is probable that multimodality therapy will be necessary for this disease. We have, therefore, tested a histone deacetylase (HDAC) inhibitor, trichostatin A (TSA), for efficacy in reducing endothelial activation. We found that pulmonary vascular endothelial VCAM-1 and tissue factor (TF) expression (both are indicators of endothelial activation) are powerfully and significantly inhibited by TSA. This is seen both with pretreatment before the inducing stress of hypoxia/reoxygenation (NY1DD sickle transgenic mouse), and upon longer-term therapy after endothelial activation has already occurred (hBERK1 sickle mouse at ambient air). In addition, TSA prevented vascular stasis in sickle mice, it exhibited activity as an iron chelator, and it induced expression of the antisickling hemoglobin, hemoglobin F. Notably, the TSA analog SAHA (suberoylanilide hydroxaminc acid) that is already approved for human clinical use exhibits the same spectrum of biologic effects as TSA. We suggest that SAHA possibly could provide true, multimodality, salubrious effects for prevention and treatment of the chronic vasculopathy of sickle cell anemia.


Subject(s)
Anemia, Sickle Cell/drug therapy , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Enzyme Inhibitors/pharmacology , Fetal Hemoglobin/genetics , Hemoglobin A/genetics , Hemoglobin, Sickle/genetics , Humans , Intercellular Adhesion Molecule-1/metabolism , Iron Chelating Agents/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pulmonary Veins/cytology , Regional Blood Flow/drug effects , Regional Blood Flow/physiology , Thromboplastin/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Venules/cytology , Venules/physiology , Vorinostat , beta-Thalassemia/drug therapy , beta-Thalassemia/genetics , beta-Thalassemia/metabolism
15.
Am J Hematol ; 85(1): 41-5, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20029945

ABSTRACT

Activation of the coagulation system is a characteristic feature of sickle cell anemia, which also includes clinical thrombosis. The sickle transgenic mouse abnormally expresses tissue factor (TF) on the pulmonary vein endothelium. Knowing that this aberrancy is stimulated by inflammation, we sought to determine whether nitric oxide (NO) contributes to regulation of endothelial TF expression in the sickle mouse model. We used the NY1DD sickle mouse, which exhibits a low-TF to high-TF phenotype switch on exposure to hypoxia/reoxygenation. Manipulations of NO biology, such as breathing NO or addition of arginine or L-NAME (N-nitro-L-arginine-methyl-ester) to the diet, caused significant modulations of TF expression. This was also seen in hBERK1 sickle mice, which have a different genetic background and already have high-TF even at ambient air. Study of NY1DD animals bred to overexpress endothelial nitric oxide synthase (eNOS; eNOS-Tg) or to have an eNOS knockout state (one eNOS(-/-) animal and several eNOS(+/-) animals) demonstrated that eNOS modulates endothelial TF expression in vivo by down-regulating it. Thus, the biodeficiency of NO characteristic of patients with sickle cell anemia may heighten risk for activation of the coagulation system.


Subject(s)
Anemia, Sickle Cell/metabolism , Endothelial Cells/metabolism , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Thromboplastin/metabolism , Animals , Cells, Cultured , Mice , Mice, Knockout , Mice, Transgenic
16.
Am J Physiol Endocrinol Metab ; 298(3): E524-33, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19996387

ABSTRACT

Gonadotropin-releasing hormone-1 (GnRH1) controls reproduction by stimulating the release of gonadotropins from the pituitary. To characterize regulatory factors governing GnRH1 gene expression, we employed biochemical and bioinformatics techniques to identify novel GnRH1 promoter-binding proteins from the brain of the cichlid fish, Astatotilapia burtoni (A. burtoni). Using an in vitro DNA-binding assay followed by mass spectrometric peptide mapping, we identified two members of the purine-rich element-binding (Pur) protein family, Puralpha and Purbeta, as candidates for GnRH1 promoter binding and regulation. We found that transcripts for both Puralpha and Purbeta colocalize in GnRH1-expressing neurons in the preoptic area of the hypothalamus in A. burtoni brain. Furthermore, we confirmed in vivo binding of endogenous Puralpha and Purbeta to the upstream region of the GnRH1 gene in A. burtoni brain and mouse neuronal GT1-7 cells. Consistent with the relative promoter occupancy exhibited by endogenous Pur proteins, overexpression of Purbeta, but not Puralpha, significantly downregulated GnRH1 mRNA levels in transiently transfected GT1-7 cells, suggesting that Purbeta acts as a repressor of GnRH1 gene transcription.


Subject(s)
Brain/physiology , Cichlids/physiology , DNA-Binding Proteins/metabolism , Gene Expression Regulation/physiology , Gonadotropin-Releasing Hormone/metabolism , Protein Precursors/metabolism , Transcriptional Activation/genetics , Animals , Mice , Nerve Tissue Proteins/metabolism
17.
Dev Cell ; 17(5): 662-73, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19922871

ABSTRACT

Myosin is the primary regulator of muscle strength and contractility. Here we show that three myosin genes, Myh6, Myh7, and Myh7b, encode related intronic microRNAs (miRNAs), which, in turn, control muscle myosin content, myofiber identity, and muscle performance. Within the adult heart, the Myh6 gene, encoding a fast myosin, coexpresses miR-208a, which regulates the expression of two slow myosins and their intronic miRNAs, Myh7/miR-208b and Myh7b/miR-499, respectively. miR-208b and miR-499 play redundant roles in the specification of muscle fiber identity by activating slow and repressing fast myofiber gene programs. The actions of these miRNAs are mediated in part by a collection of transcriptional repressors of slow myofiber genes. These findings reveal that myosin genes not only encode the major contractile proteins of muscle, but act more broadly to influence muscle function by encoding a network of intronic miRNAs that control muscle gene expression and performance.


Subject(s)
Gene Expression Regulation, Developmental , MicroRNAs/genetics , Muscle, Skeletal/metabolism , Myosin Heavy Chains/genetics , Animals , Base Sequence , Cardiac Myosins/genetics , Cardiac Myosins/metabolism , Cell Line , Chlorocebus aethiops , Mice , Myosin Heavy Chains/metabolism , Myosin Type II/genetics , Myosin Type II/metabolism
18.
J Biol Chem ; 284(47): 32312-20, 2009 Nov 20.
Article in English | MEDLINE | ID: mdl-19720825

ABSTRACT

Expression of the arginine/lysine transporter Cat-1 is highly induced in proliferating and stressed cells via mechanisms that include transcriptional activation. A bifunctional INE (intronic element) within the first intron of the Cat-1 gene was identified and characterized in this study. The INE had high sequence homology to an amino acid response element and was shown to act as a transcriptional enhancer in unstressed cells by binding the transcription factor, purine-rich element binding protein A (Pur alpha). During endoplasmic reticulum stress, binding of Pur alpha to the INE decreased; the element acted as a positive regulator in early stress by binding of the transcription factor ATF4 and as a negative regulator in prolonged stress by binding the stress-induced C/EBP family member, CHOP. We conclude that transcriptional control of the Cat-1 gene is tightly controlled by multiple cis-DNA elements, contributing to regulation of cationic amino acid transport for cell growth and proliferation. In addition, we propose that genes may use stress-response elements such as the INE to support basal expression in the absence of stress.


Subject(s)
Cationic Amino Acid Transporter 1/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation , Nerve Tissue Proteins/genetics , Transcription Factors/genetics , Activating Transcription Factor 4/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , DNA/chemistry , Endoplasmic Reticulum/metabolism , Enhancer Elements, Genetic , Humans , Introns , Mice , Rats , Transcription Factor CHOP/metabolism
19.
Biochemistry ; 48(27): 6348-60, 2009 Jul 14.
Article in English | MEDLINE | ID: mdl-19496623

ABSTRACT

Expression of the smooth muscle alpha-actin gene in growth-activated vascular smooth muscle cells and stromal fibroblasts is negatively regulated by members of the Pur family of single-stranded DNA/RNA-binding proteins. In particular, Puralpha and Purbeta are postulated to repress transcription by forming helix-destabilizing complexes with the sense strand of an asymmetric polypurine-polypyrimidine tract containing a canonical MCAT enhancer motif in the 5' region of the gene. Herein, we establish the mechanism of Purbeta binding to the purine-rich strand of the enhancer using quantitative methods and purified components. Initial evaluation of DNA-binding specificity and equilibrium stoichiometry via colorimetric-, autoradiographic-, and fluorescence-based assays suggested that Purbeta interacts with two distinct G/A-rich sites within the nominal single-stranded enhancer element to form a high-affinity 2:1 protein:DNA complex. Statistical mechanical analyses of band shift titrations of the nominal element in conjunction with DNase I footprint titrations of the extended smooth muscle alpha-actin 5'-flanking region demonstrated that assembly of the nucleoprotein complex likely occurs in a sequential, cooperative, and monomer-dependent fashion. Resolution of the microscopic energetics of the system indicated that monomer association with two nonidentical sites flanking the core MCAT motif accounts for the majority of the intrinsic binding affinity of Purbeta with intersite cooperativity contributing an approximately 12-fold increase to the stability of the nucleoprotein complex. These findings offer new insights into the mechanism, energetics, and sequence determinants of Purbeta repressor binding to a biologically relevant, contractile phenotype-regulating cis-element while also revealing the thermodynamic confines of putative Purbeta-mediated effects on DNA structure.


Subject(s)
Actins/genetics , DNA-Binding Proteins/chemistry , Enhancer Elements, Genetic , Muscle, Smooth/chemistry , Adenosine Triphosphate/chemistry , Base Sequence , Colorimetry , DNA Footprinting , DNA Primers , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Fluorescence Polarization
20.
Mol Biol Cell ; 20(8): 2174-85, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19261809

ABSTRACT

Transforming growth factor (TGF) beta1 is a mediator of myofibroblast differentiation in healing wounds in which it activates transcription of the smooth muscle alpha-actin (SMalphaA) gene via dynamic interplay of nuclear activators and repressors. Targeting components of TGFbeta1 signaling may be an effective strategy for controlling myofibroblasts in chronic fibrotic diseases. We examined the ability of proinflammatory tumor necrosis factor (TNF)-alpha to antagonize TGFbeta1-mediated human pulmonary myofibroblast differentiation. TNF-alpha abrogated TGFbeta1-induced SMalphaA gene expression at the level of transcription without disrupting phosphorylation of regulatory Smads. Intact mitogen-activated protein kinase kinase (Mek)-extracellular signal-regulated kinase (Erk) kinase signaling was required for myofibroblast repression by TNF-alpha via induction of the early growth response factor-1 (Egr-1) DNA-binding protein. Egr-1 bound to the GC-rich SPUR activation element in the SMalphaA promoter and potently suppressed Smad3- and TGFbeta1-mediated transcription. Reduction in Smad binding to the SMalphaA promoter in TNF-alpha-treated myofibroblasts was accompanied by an increase in Egr-1 and YB-1 repressor binding, suggesting that the molecular mechanism underlying repression may involve competitive interplay between Egr-1, YB-1, and Smads. The ability of TNF-alpha to attenuate myofibroblast differentiation via modulation of a Mek1/Erk/Egr-1 regulatory axis may be useful in designing new therapeutic targets to offset destructive tissue remodeling in chronic fibrotic disease.


Subject(s)
Actins/genetics , Early Growth Response Protein 1/metabolism , Fibroblasts/enzymology , Gene Expression Regulation/drug effects , MAP Kinase Kinase 1/metabolism , Transforming Growth Factor beta1/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/drug effects , Humans , Lung/cytology , Models, Genetic , Muscle, Smooth/metabolism , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Repressor Proteins/metabolism , Smad2 Protein/metabolism , Smad7 Protein/metabolism , Transcription, Genetic/drug effects , Y-Box-Binding Protein 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...