Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
J Biol Chem ; 299(3): 102912, 2023 03.
Article in English | MEDLINE | ID: mdl-36649910

ABSTRACT

Daptomycin (DAP) is an antibiotic frequently used as a drug of last resort against vancomycin-resistant enterococci. One of the major challenges when using DAP against vancomycin-resistant enterococci is the emergence of resistance, which is mediated by the cell-envelope stress system LiaFSR. Indeed, inhibition of LiaFSR signaling has been suggested as a strategy to "resensitize" enterococci to DAP. In the absence of LiaFSR, alternative pathways mediating DAP resistance have been identified, including adaptive mutations in the enolpyruvate transferase MurAA (MurAAA149E), which catalyzes the first committed step in peptidoglycan biosynthesis; however, how these mutations confer resistance is unclear. Here, we investigated the biochemical basis for MurAAA149E-mediated adaptation to DAP to determine whether such an alternative pathway would undermine the potential efficacy of therapies that target the LiaFSR pathway. We found cells expressing MurAAA149E had increased susceptibility to glycoside hydrolases, consistent with decreased cell wall integrity. Furthermore, structure-function studies of MurAA and MurAAA149E using X-ray crystallography and biochemical analyses indicated only a modest decrease in MurAAA149E activity, but a 16-fold increase in affinity for MurG, which performs the last intracellular step of peptidoglycan synthesis. Exposure to DAP leads to mislocalization of cell division proteins including MurG. In Bacillus subtilis, MurAA and MurG colocalize at division septa and, thus, we propose MurAAA149E may contribute to DAP nonsusceptibility by increasing the stability of MurAA-MurG interactions to reduce DAP-induced mislocalization of these essential protein complexes.


Subject(s)
Daptomycin , Enterococcus faecium , Transferases , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Bacterial Proteins/metabolism , Daptomycin/metabolism , Daptomycin/pharmacology , Drug Resistance, Bacterial , Enterococcus faecium/drug effects , Enterococcus faecium/metabolism , Microbial Sensitivity Tests , Peptidoglycan/metabolism , Transferases/metabolism
2.
Methods ; 90: 28-38, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26344758

ABSTRACT

Calcium signaling is a ubiquitous and versatile process involved in nearly every cellular process, and exploitation of host calcium signals is a common strategy used by viruses to facilitate replication and cause disease. Small molecule fluorescent calcium dyes have been used by many to examine changes in host cell calcium signaling and calcium channel activation during virus infections, but disadvantages of these dyes, including poor loading and poor long-term retention, complicate analysis of calcium imaging in virus-infected cells due to changes in cell physiology and membrane integrity. The recent expansion of genetically-encoded calcium indicators (GECIs), including blue and red-shifted color variants and variants with calcium affinities appropriate for calcium storage organelles like the endoplasmic reticulum (ER), make the use of GECIs an attractive alternative for calcium imaging in the context of virus infections. Here we describe the development and testing of cell lines stably expressing both green cytoplasmic (GCaMP5G and GCaMP6s) and red ER-targeted (RCEPIAer) GECIs. Using three viruses (rotavirus, poliovirus and respiratory syncytial virus) previously shown to disrupt host calcium homeostasis, we show the GECI cell lines can be used to detect simultaneous cytoplasmic and ER calcium signals. Further, we demonstrate the GECI expression has sufficient stability to enable long-term confocal imaging of both cytoplasmic and ER calcium during the course of virus infections.


Subject(s)
Calcium Signaling , Calcium/analysis , Host-Pathogen Interactions , Microscopy, Fluorescence/methods , Animals , Cell Line , Chlorocebus aethiops , Endoplasmic Reticulum/metabolism , HEK293 Cells , Humans , Indicators and Reagents/chemistry , Poliovirus/metabolism , Respiratory Syncytial Viruses/metabolism , Rotavirus/metabolism
3.
Proc Natl Acad Sci U S A ; 109(50): E3405-13, 2012 Dec 11.
Article in English | MEDLINE | ID: mdl-23184977

ABSTRACT

Autophagy is a cellular degradation process involving an intracellular membrane trafficking pathway that recycles cellular components or eliminates intracellular microbes in lysosomes. Many pathogens subvert autophagy to enhance their replication, but the mechanisms these pathogens use to initiate the autophagy process have not been elucidated. This study identifies rotavirus as a pathogen that encodes a viroporin, nonstructural protein 4, which releases endoplasmic reticulum calcium into the cytoplasm, thereby activating a calcium/calmodulin-dependent kinase kinase-ß and 5' adenosine monophosphate-activated protein kinase-dependent signaling pathway to initiate autophagy. Rotavirus hijacks this membrane trafficking pathway to transport viral proteins from the endoplasmic reticulum to sites of viral replication to produce infectious virus. This process requires PI3K activity and autophagy-initiation proteins Atg3 and Atg5, and it is abrogated by chelating cytoplasmic calcium or inhibiting calcium/calmodulin-dependent kinase kinase-ß. Although the early stages of autophagy are initiated, rotavirus infection also blocks autophagy maturation. These studies identify a unique mechanism of virus-mediated, calcium-activated signaling that initiates autophagy and hijacks this membrane trafficking pathway to transport viral proteins to sites of viral assembly.


Subject(s)
Autophagy/physiology , Calcium-Calmodulin-Dependent Protein Kinase Kinase/physiology , Rotavirus/physiology , Virus Replication/physiology , Animals , Autophagy-Related Protein 5 , Autophagy-Related Proteins , Calcium Signaling , Cell Line , Cells, Cultured , Enzyme Activation , Glycoproteins/physiology , Macaca mulatta , Mice , Mice, Knockout , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/physiology , Protein Transport , Rotavirus/pathogenicity , Signal Transduction , Toxins, Biological/physiology , Ubiquitin-Conjugating Enzymes/deficiency , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/physiology , Unfolded Protein Response , Viral Nonstructural Proteins/physiology , Virus Assembly/physiology
4.
J Virol ; 87(24): 13579-88, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24109210

ABSTRACT

Rotavirus nonstructural protein 4 (NSP4) induces dramatic changes in cellular calcium homeostasis. These include increased endoplasmic reticulum (ER) permeability, resulting in decreased ER calcium stores and activation of plasma membrane (PM) calcium influx channels, ultimately causing a 2- to 4-fold elevation in cytoplasmic calcium. Elevated cytoplasmic calcium is absolutely required for virus replication, but the underlying mechanisms responsible for calcium influx remain poorly understood. NSP4 is an ER-localized viroporin, whose activity depletes ER calcium, which ultimately leads to calcium influx. We hypothesized that NSP4-mediated depletion of ER calcium activates store-operated calcium entry (SOCE) through activation of the ER calcium sensor stromal interaction molecule 1 (STIM1). We established and used a stable yellow fluorescent protein-expressing STIM1 cell line (YFP-STIM1) as a biosensor to assess STIM1 activation (puncta formation) by rotavirus infection and NSP4 expression. We found that STIM1 is constitutively active in rotavirus-infected cells and that STIM1 puncta colocalize with the PM-localized Orai1 SOCE calcium channel. Expression of wild-type NSP4 activated STIM1, resulting in PM calcium influx, but an NSP4 viroporin mutant failed to induce STIM1 activation and did not activate the PM calcium entry pathway. Finally, knockdown of STIM1 significantly reduced rotavirus yield, indicating STIM1 plays a critical role in virus replication. These data demonstrate that while rotavirus may ultimately activate multiple calcium channels in the PM, calcium influx is predicated on NSP4 viroporin-mediated activation of STIM1 in the ER. This is the first report of viroporin-mediated activation of SOCE, reinforcing NSP4 as a robust model to understand dysregulation of calcium homeostasis during virus infections.


Subject(s)
Calcium/metabolism , Endoplasmic Reticulum/metabolism , Glycoproteins/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Rotavirus Infections/metabolism , Rotavirus Infections/virology , Rotavirus/metabolism , Toxins, Biological/metabolism , Viral Nonstructural Proteins/metabolism , Cell Line , Cell Membrane/metabolism , Cytoplasm/metabolism , Endoplasmic Reticulum/genetics , Glycoproteins/genetics , Humans , Ion Transport , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Rotavirus/genetics , Rotavirus Infections/genetics , Stromal Interaction Molecule 1 , Toxins, Biological/genetics , Viral Nonstructural Proteins/genetics
5.
J Virol ; 86(9): 4921-34, 2012 May.
Article in English | MEDLINE | ID: mdl-22357281

ABSTRACT

Nonstructural protein 4 (NSP4) viroporin activity is critical for the replication and assembly of serogroup A rotavirus (RVA); however, the dramatic primary sequence divergence of NSP4s across serogroups raises the possibility that viroporin activity is not a common feature among RVs. We tested for NSP4 viroporin activity from divergent strains, including RVA (EC and Ty-1), RVB (IDIR), and RVC (Cowden). Canonical viroporin motifs were identified in RVA, RVB, and RVC NSP4s, but the arrangement of basic residues and the amphipathic α-helices was substantially different between serogroups. Using Escherichia coli and mammalian cell expression, we showed that each NSP4 tested had viroporin activity, but serogroup-specific viroporin phenotypes were identified. Only mammalian RVA and RVC NSP4s induced BL21-pLysS E. coli cell lysis, a classical viroporin activity assay. In contrast, RVA, RVB, and RVC NSP4 expression was universally cytotoxic to E. coli and disrupted reduction-oxidation activities, as measured by a new redox dye assay. In mammalian cells, RVB and RVC NSP4s were initially localized in the endoplasmic reticulum (ER) and trafficked into punctate structures that were mutually exclusive with RVA NSP4. The punctate structures partially localized to the ER-Golgi intermediate compartment (ERGIC) but primarily colocalized with punctate LC3, a marker for autophagosomes. Similar to RVA NSP4, expression of RVB and RVC NSP4s significantly elevated cytosolic calcium levels, demonstrating that despite strong primary sequence divergence, RV NSP4 has maintained viroporin activity across serogroups A to C. These data suggest that elevated cytosolic calcium is a common critical process for all rotavirus strains.


Subject(s)
Glycoproteins/genetics , Glycoproteins/metabolism , Rotavirus/genetics , Rotavirus/metabolism , Toxins, Biological/genetics , Toxins, Biological/metabolism , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Amino Acid Sequence , Animals , Calcium/metabolism , Chlorocebus aethiops , Escherichia coli/metabolism , Escherichia coli/virology , Glycoproteins/immunology , Intracellular Space/metabolism , Molecular Sequence Data , Oxidation-Reduction , Protein Structure, Tertiary , Protein Transport , Rotavirus/classification , Sequence Homology, Amino Acid , Toxins, Biological/immunology , Viral Nonstructural Proteins/immunology
6.
Sci Rep ; 13(1): 5813, 2023 04 10.
Article in English | MEDLINE | ID: mdl-37037845

ABSTRACT

Half of the marine virosphere is hypothesized to be RNA viruses (kingdom Orthornavirae) that infect abundant micro-eukaryotic hosts (e.g. protists). To test this, quantitative approaches that broadly track infections in situ are needed. Here, we describe a technique-dsRNA-Immunofluorescence (dsRIF)-that uses a double-stranded RNA (dsRNA) targeting monoclonal antibody to assess host infection status based on the presence of dsRNA, a replicative intermediate of all Orthornavirae infections. We show that the dinoflagellate Heterocapsa circularisquama produces dsRIF signal ~ 1000 times above background autofluorescence when infected by the + ssRNA virus HcRNAV. dsRNA-positive virocells were detected across > 50% of the 48-h infection cycle and accumulated to represent at least 63% of the population. Photosynthetic and chromosomal integrity remained intact during peak replication, indicating HcRNAV infection does not interrupt these processes. This work validates the use of dsRIF on marine RNA viruses and their hosts, setting the stage for quantitative environmental applications that will accelerate understanding of virus-driven ecosystem impacts.


Subject(s)
Dinoflagellida , RNA Virus Infections , RNA Viruses , Viruses , Humans , RNA, Viral/genetics , Ecosystem , RNA Viruses/genetics , Viruses/genetics , Dinoflagellida/genetics , RNA, Double-Stranded
7.
Virol J ; 9: 181, 2012 Sep 03.
Article in English | MEDLINE | ID: mdl-22943503

ABSTRACT

BACKGROUND: Our previous report that the Norwalk virus nonstructural protein p22 is an antagonist of the cellular secretory pathway suggests a new aspect of norovirus/host interaction. To explore conservation of function of this highly divergent calicivirus protein, we examined the effects of p22 homologues from four human and two murine noroviruses, and feline calicivirus on the secretory pathway. FINDINGS: All human noroviruses examined induced Golgi disruption and inhibited protein secretion, with the genogroup II.4 Houston virus being the most potent antagonist. Genogroup II.6 viruses have a conserved mutation in the mimic of an Endoplasmic Reticulum export signal (MERES) motif that is highly conserved in human norovirus homologues of p22 and is critical for secretory pathway antagonism, and these viruses had reduced levels of Golgi disruption and inhibition of protein secretion. p22 homologues from both persistent and nonpersistent strains of murine norovirus induced Golgi disruption, but only mildly inhibited cellular protein secretion. Feline calicivirus p30 did not induce Golgi disruption or inhibit cellular protein secretion. CONCLUSIONS: These differences confirm a norovirus-specific effect on host cell secretory pathway antagonism by homologues of p22, which may affect viral replication and/or cellular pathogenesis.


Subject(s)
Host-Pathogen Interactions , Norwalk virus/genetics , Norwalk virus/pathogenicity , Secretory Pathway , Viral Nonstructural Proteins/metabolism , Virulence Factors/metabolism , Amino Acid Sequence , Animals , Cell Line , Golgi Apparatus/virology , Humans , Molecular Sequence Data , Sequence Alignment , Viral Nonstructural Proteins/genetics , Virulence Factors/genetics
8.
Nat Commun ; 13(1): 3057, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35650195

ABSTRACT

Desmoplastic small round cell tumor (DSRCT) is an aggressive, usually incurable sarcoma subtype that predominantly occurs in post-pubertal young males. Recent evidence suggests that the androgen receptor (AR) can promote tumor progression in DSRCTs. However, the mechanism of AR-induced oncogenic stimulation remains undetermined. Herein, we demonstrate that enzalutamide and AR-directed antisense oligonucleotides (AR-ASO) block 5α-dihydrotestosterone (DHT)-induced DSRCT cell proliferation and reduce xenograft tumor burden. Gene expression analysis and chromatin immunoprecipitation sequencing (ChIP-seq) were performed to elucidate how AR signaling regulates cellular epigenetic programs. Remarkably, ChIP-seq revealed novel DSRCT-specific AR DNA binding sites adjacent to key oncogenic regulators, including WT1 (the C-terminal partner of the pathognomonic fusion protein) and FOXF1. Additionally, AR occupied enhancer sites that regulate the Wnt pathway, neural differentiation, and embryonic organ development, implicating AR in dysfunctional cell lineage commitment. Our findings have direct clinical implications given the widespread availability of FDA-approved androgen-targeted agents used for prostate cancer.


Subject(s)
Androgen Receptor Antagonists , Desmoplastic Small Round Cell Tumor , Receptors, Androgen , Androgen Receptor Antagonists/pharmacology , Androgens , Animals , Cell Line, Tumor , Desmoplastic Small Round Cell Tumor/genetics , Humans , Male , Oligonucleotides, Antisense/pharmacology , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Xenograft Model Antitumor Assays
9.
Proc Natl Acad Sci U S A ; 105(26): 8811-8, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18587047

ABSTRACT

Rotavirus NSP4 is a viral enterotoxin capable of causing diarrhea in neonatal mice. This process is initiated by the binding of extracellular NSP4 to target molecule(s) on the cell surface that triggers a signaling cascade leading to diarrhea. We now report that the integrins alpha1beta1 and alpha2beta1 are receptors for NSP4. NSP4 specifically binds to the alpha1 and alpha2 I domains with apparent K(d) = 1-2.7 muM. Binding is mediated by the I domain metal ion-dependent adhesion site motif, requires Mg(2+) or Mn(2+), is abolished with EDTA, and an NSP4 point mutant, E(120)A, fails to bind alpha2 integrin I domain. NSP4 has two distinct integrin interaction domains. NSP4 amino acids 114-130 are essential for binding to the I domain, and NSP4 peptide 114-135 blocks binding of the natural ligand, collagen I, to integrin alpha2. NSP4 amino acids 131-140 are not associated with the initial binding to the I domain, but elicit signaling that leads to the spreading of attached C2C12-alpha2 cells, mouse myoblast cells stably expressing the human alpha2 integrin. NSP4 colocalizes with integrin alpha2 on the basolateral surface of rotavirus-infected polarized intestinal epithelial (Caco-2) cells as well as surrounding noninfected cells. NSP4 mutants that fail to bind or signal through integrin alpha2 were attenuated in diarrhea induction in neonatal mice. These results indicate that NSP4 interaction with integrin alpha1 and alpha2 is an important component of enterotoxin function and rotavirus pathogenesis, further distinguishing this viral virulence factor from other microbial enterotoxins.


Subject(s)
Enterotoxins/metabolism , Glycoproteins/metabolism , Integrin alpha1beta1/metabolism , Integrin alpha2beta1/metabolism , Rotavirus/metabolism , Toxins, Biological/metabolism , Viral Nonstructural Proteins/metabolism , Androstadienes/pharmacology , Animals , Binding Sites , Cell Adhesion/drug effects , Cell Line , Cell Movement/drug effects , Diarrhea/chemically induced , Diarrhea/metabolism , Enterotoxins/chemistry , Enzyme-Linked Immunosorbent Assay , Estrenes/pharmacology , Glycoproteins/chemistry , Humans , Integrin alpha1beta1/chemistry , Integrin alpha2beta1/chemistry , Mice , Protein Binding/drug effects , Protein Structure, Tertiary , Protein Transport/drug effects , Pyrrolidinones/pharmacology , Rotavirus/drug effects , Signal Transduction/drug effects , Surface Plasmon Resonance , Toxins, Biological/chemistry , Viral Nonstructural Proteins/chemistry , Wortmannin
10.
Cancer Gene Ther ; 28(12): 1325-1338, 2021 12.
Article in English | MEDLINE | ID: mdl-33408328

ABSTRACT

Osteosarcoma (OS) is a molecularly heterogeneous, aggressive, poorly differentiated pediatric bone cancer that frequently spreads to the lung. Relatively little is known about phenotypic and epigenetic changes that promote lung metastases. To identify key drivers of metastasis, we studied human CCH-OS-D OS cells within a previously described rat acellular lung (ACL) model that preserves the native lung architecture, extracellular matrix, and capillary network. This system identified a subset of cells-termed derived circulating tumor cells (dCTCs)-that can migrate, intravasate, and spread within a bioreactor-perfused capillary network. Remarkably, dCTCs highly expressed epithelial-to-mesenchymal transition (EMT)-associated transcription factors (EMT-TFs), such as ZEB1, TWIST, and SOX9, which suggests that they undergo cellular reprogramming toward a less differentiated state by coopting the same epigenetic machinery used by carcinomas. Since YAP/TAZ and AXL tightly regulate the fate and plasticity of normal mesenchymal cells in response to microenvironmental cues, we explored whether these proteins contributed to OS metastatic potential using an isogenic pair of human OS cell lines that differ in AXL expression. We show that AXL inhibition significantly reduced the number of MG63.2 pulmonary metastases in murine models. Collectively, we present a laboratory-based method to detect and characterize a pure population of dCTCs, which provides a unique opportunity to study how OS cell fate and differentiation contributes to metastatic potential. Though the important step of clinical validation remains, our identification of AXL, ZEB1, and TWIST upregulation raises the tantalizing prospect that EMT-TF-directed therapies might expand the arsenal of therapies used to combat advanced-stage OS.


Subject(s)
Osteosarcoma/genetics , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , YAP-Signaling Proteins/metabolism , Animals , Cell Dedifferentiation , Disease Models, Animal , Humans , Mice , Neoplasm Metastasis , Osteosarcoma/pathology , Axl Receptor Tyrosine Kinase
11.
Circulation ; 114(1 Suppl): I200-5, 2006 Jul 04.
Article in English | MEDLINE | ID: mdl-16820572

ABSTRACT

BACKGROUND: Thoracic aortic dissection (TAD) is characterized by dysregulated extracellular matrix. Little is known about the alterations of collagen and stimulators of collagen synthesis, eg, connective tissue growth factor (CTGF), in patients with TAD. In this study, we examined their roles in TAD. METHODS AND RESULTS: Surgical specimens of the aortic wall of TAD patients (n=10) and controls (n=10) were tested for collagen types I and III and CTGF expression. When compared with controls, protein levels of type I and III collagen and CTGF were significantly increased by 3.2-, 3.7-, and 5.3-fold, respectively (P<0.05 for all). Similar patterns were shown in mRNA levels of type Ialpha and Ialpha2 collagen and CTGF. Using immunohistochemistry and trichrome staining, we also observed elevated levels of collagen in the aortic media and adventitia. Treatment with recombinant human CTGF increased collagen synthesis in cultured aortic smooth muscle cells in a dose- and time-dependent fashion, in which expression of collagens increased from 506+/-108 counts per minute to 2764+/-240 cpm by 50 ng/mL CTGF, and from 30+/-43 cpm to 429+/-102 cpm at 48 hours. CONCLUSIONS: TAD patients exhibited significantly increased expression of aortic collagen types I and III as well as CTGF, which is likely to be responsible for the compromised aortic distensibility and systemic compliance. Because CTGF can increase collagen expression, CTGF may be a new target molecule in the pathogenesis and progression of TAD.


Subject(s)
Aortic Aneurysm, Thoracic/metabolism , Aortic Dissection/metabolism , Collagen Type III/biosynthesis , Collagen Type I/biosynthesis , Immediate-Early Proteins/physiology , Intercellular Signaling Peptides and Proteins/physiology , Adult , Aged , Aortic Dissection/genetics , Aortic Dissection/pathology , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Aortic Aneurysm, Thoracic/genetics , Aortic Aneurysm, Thoracic/pathology , Apoptosis , Collagen Type I/genetics , Collagen Type III/genetics , Compliance , Connective Tissue Growth Factor , Female , Gene Expression Profiling , Humans , Immediate-Early Proteins/biosynthesis , Immediate-Early Proteins/genetics , Immediate-Early Proteins/pharmacology , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Male , Middle Aged , Muscle, Smooth, Vascular/pathology , Recombinant Proteins/pharmacology
12.
Cardiovasc Res ; 69(2): 502-11, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16316638

ABSTRACT

OBJECTIVES: Atherosclerosis is the leading cause of death in the United States, and human cytomegalovirus (HCMV) infection may play a role in the development of this disease. Diminished expression and/or activity of endothelial nitric oxide synthase (eNOS) are an early event in atherogenesis. In the current study, we investigated the effects of HCMV infection on eNOS activation in human aortic endothelial cells (HAECs). METHODS AND RESULTS: We found that HCMV inhibited eNOS phosphorylation/activation in HAECs. The signaling upstream of eNOS involving Akt and PDK1 were also suppressed by the HCMV infection. Moreover, HCMV infection increased the expression of PTEN (phosphatase and tensin homolog deleted on chromosome 10). Silencing PTEN expression with specific siRNA reversed the inhibitory effects on eNOS activation in HCMV-infected cells indicating the involvement of PTEN in mediating HCMV's inhibitory effects. Next we observed that the activation of p38 MAPK stress signaling pathway mediates HCMV's effects on PTEN up-regulation and eNOS inactivation. CONCLUSIONS: In summary, our findings suggest that inhibition of eNOS leading to endothelial dysfunction may be a basis of the pro-atherogenic effects of HCMV. Importantly, upregulation of PTEN and activation of stress signal p38 MAPK are involved in HCMV's inhibitory effects on eNOS activation.


Subject(s)
Cytomegalovirus Infections/metabolism , Cytomegalovirus , Endothelium, Vascular/metabolism , Nitric Oxide Synthase Type III/metabolism , PTEN Phosphohydrolase/metabolism , Up-Regulation , 3-Phosphoinositide-Dependent Protein Kinases , Atherosclerosis/metabolism , Blotting, Western/methods , Cells, Cultured , Endothelial Cells , Enzyme Activation , Fluorescent Antibody Technique , Humans , MAP Kinase Signaling System , Oncogene Protein v-akt/metabolism , PTEN Phosphohydrolase/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/genetics , Transfection , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Sci Rep ; 7: 44667, 2017 03 20.
Article in English | MEDLINE | ID: mdl-28317850

ABSTRACT

Mammalian parental genomes contribute differently to early embryonic development. Before activation of the zygotic genome, the maternal genome provides all transcripts and proteins required for the transition from a highly specialized oocyte to a pluripotent embryo. Depletion of these maternally-encoded transcripts frequently results in failure of preimplantation embryonic development, but their functions in this process are incompletely understood. We found that female mice lacking NLRP2 are subfertile because of early embryonic loss and the production of fewer offspring that have a wide array of developmental phenotypes and abnormal DNA methylation at imprinted loci. By demonstrating that NLRP2 is a member of the subcortical maternal complex (SCMC), an essential cytoplasmic complex in oocytes and preimplantation embryos with poorly understood function, we identified imprinted postzygotic DNA methylation maintenance, likely by directing subcellular localization of proteins involved in this process, such as DNMT1, as a new crucial role of the SCMC for mammalian reproduction.


Subject(s)
Cellular Reprogramming/genetics , Embryonic Development , Epigenesis, Genetic , Fertility , Multiprotein Complexes/metabolism , Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Apoptosis Regulatory Proteins , Blastocyst/metabolism , Cell Shape , DNA Methylation/genetics , Embryo, Mammalian/metabolism , Embryonic Development/genetics , Female , Fertility/genetics , Fertilization/genetics , Genomic Imprinting , Male , Mice , Oocytes/cytology , Oocytes/metabolism , Pregnancy , Pregnancy Outcome , Protein Binding
14.
FEBS Lett ; 580(11): 2779-87, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16650413

ABSTRACT

Human cytomegalovirus (HCMV) exerts anti-apoptotic effect during early stage of infection, which provides HCMV time for propagation. We investigated pathways mediating the resistance to H(2)O(2)-induced cell death - a self-defense mechanism to remove infected cells. We found that human aortic endothelial cells (HAECs) infected with VHL/E strain of HCMV during first 3 days were resistant to H(2)O(2) (0-2 mM) induced apoptosis. This anti-apoptotic effect may be mediated by the upregulation of Bcl-2, an anti-apoptotic protein through the activation pro-survival pathway extracellular signal regulated kinase (ERK). Through this mechanism, HCMV is able to propagate and causes endothelial dysfunction, hence vascular disease.


Subject(s)
Apoptosis/drug effects , Cytomegalovirus/physiology , Endothelial Cells/drug effects , Endothelial Cells/virology , Hydrogen Peroxide/pharmacology , MAP Kinase Signaling System/drug effects , Caspase 3 , Caspases/metabolism , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/enzymology , Enzyme Activation/drug effects , Humans , Intracellular Membranes/drug effects , Membrane Potentials/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/genetics , Up-Regulation
15.
Arterioscler Thromb Vasc Biol ; 25(12): 2509-14, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16210567

ABSTRACT

BACKGROUND: Previously, we showed that the 27nt repeat polymorphism in endothelial nitric oxide synthase (eNOS) intron 4 was associated with altered eNOS mRNA and protein levels, nitric oxide (NO) production and vascular disease risk; the 27-nt repeats had a cis-acting role in eNOS promoter function. In the present study, we investigated nuclear protein that binds the 27nt repeat and mediates eNOS expression. METHODS AND RESULTS: Using 5'-biotin-labeled 27nt DNA duplex and streptavidin-agarose beads pull-down assay and mass spectrometry, we identified that nuclear beta-actin was one of the major 27nt binding proteins. Using the pGL3 reporter vectors containing the 5x27nt repeats as an enhancer in an in vitro transcription assay, we found that exogenous beta-actin significantly increased reporter gene transcription efficiency. The beta-actin's upregulating effect was compromised when exogenous 27nt RNA duplex was added. Furthermore, the eNOS expression was reduced when beta-actin gene was silenced by specific siRNA, and actin overexpression upregulated eNOS expression >3-fold. CONCLUSIONS: Our data demonstrate that beta-actin as a transcription factor stimulates eNOS expression; and the transcriptional effect appears to be 27nt-dependent. Our findings represent a novel molecular mechanism regulating eNOS expression, which could potentially lead to discoveries of eNOS specific pharmaceutical agents, eg, active peptides, with clinical applications.


Subject(s)
Actins/metabolism , Endothelium, Vascular/enzymology , Gene Expression Regulation, Enzymologic/physiology , Nitric Oxide Synthase Type III/genetics , Transcription Factors/metabolism , Actins/genetics , Aorta/cytology , Cell Nucleus/physiology , Cells, Cultured , Endothelium, Vascular/cytology , Gene Silencing , Genes, Reporter , Humans , Introns , Polymorphism, Genetic , RNA, Messenger/metabolism , Transcription Factors/genetics , Transcription, Genetic/physiology
16.
FEBS Lett ; 579(3): 733-40, 2005 Jan 31.
Article in English | MEDLINE | ID: mdl-15670837

ABSTRACT

Cigarette-induced endothelial dysfunction could be an early mediator of atherosclerosis. In this study, we explored the mechanisms of cigarette smoke extract (CSE)-induced human aortic endothelial cells (HAEC) apoptosis. We found that 10-65% of HAECs underwent apoptotic changes when HAECs were exposed to 0.001-0.02 cigarette equivalent unit of CSE for 4 h. CSE activated the caspases-3 and 8, the p38 MAP kinase and stress activated protein kinase/c-Jun N-terminal protein kinase (SAPK/JNK). Specific inhibitors of p38 MAP or SAPK/JNK reduced CSE-induced caspase activation. We further showed that eNOS pre-activation by L-arginine reduced endothelial apoptosis from 65% to 5%; and eNOS inhibition by N-omega-nitro-L-arginine methyl ester accentuated CSE-induced endothelial apoptosis. We suggest that appropriate endogenous NO production may be an important protective mechanism against smoking-induced endothelial damage.


Subject(s)
Apoptosis , Endothelium, Vascular/cytology , Nicotiana , Nitric Oxide/physiology , Smoking , Base Sequence , Blotting, Western , Caspase 3 , Caspase 8 , Caspases/metabolism , Cells, Cultured , DNA Primers , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Humans , Mitogen-Activated Protein Kinases/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III
17.
DNA Cell Biol ; 24(4): 218-24, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15812238

ABSTRACT

DNA polymorphisms in endothelial nitric oxide synthase (eNOS) gene have been shown to be associated with constitutive eNOS expression and coronary artery disease (CAD). In the present study we explored the hypothesis whether genotype-dependent effects can be maintained in vitro during replication, or the effect is conditional on in vivo biological environments. Human umbilical vein endothelial cells (HUVEC) were collected and cultured from 89 normal deliveries of Mexican Americans. The cells were treated with or without cigarette smoking extracts (CSE) and genotypes of eNOS polymorphisms were determined by PCR. We measured the levels of eNOS by ELISA and its binding proteins including heat-shock protein 90 (Hsp-90) and caveolin-1 by Western blotting. The rare C allele for the promoter T786C polymorphism (0.2), and the rare 4 x 27-bp repeat allele in the intron 4 (0.30) were different from those reported in other populations. Yet, the rare T allele in the exon 7 (G894T polymorphism) was similar as others. After four passages in vitro, both the intron 4 and promoter polymorphisms maintained significant effects on eNOS mRNA levels in HUVECs (P < 0.05). However, the effects on eNOS protein and enzyme activity were less consistent. Although primary smokers had significantly lower eNOS protein levels (P < 0.05), the in vitro CSE treatment on cultured HUVECs only resulted in a significant reduction in NO levels as measured by the stable metabolites of nitrite/nitrate (P < 0.001). Neither Hsp-90 nor caveolin-1--important eNOS regulators--appears to mediate the genotypesmoking effects on eNOS expression although HUVECs did produce more Hsp-90 when exposed to CSE. Our study demonstrates that endothelial cells maintain genotype-dependent expression even after the deprivation of in vivo environment. However, the cigarette smoking-genotype interaction may require such in vivo conditions to be manifested.


Subject(s)
Endothelial Cells/metabolism , Gene Expression/genetics , Nitric Oxide Synthase/metabolism , Polymorphism, Genetic , RNA, Messenger/metabolism , Umbilical Veins/cytology , Analysis of Variance , Blotting, Western , Caveolin 1 , Caveolins/metabolism , Cells, Cultured , DNA Primers , Enzyme-Linked Immunosorbent Assay , Gene Expression/drug effects , Genotype , HSP90 Heat-Shock Proteins/metabolism , Humans , Mexican Americans , Mutation/genetics , Nitrates/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type III , Nitrites/metabolism , Polymerase Chain Reaction , Tars/toxicity
18.
Surgery ; 138(2): 352-9, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16153447

ABSTRACT

BACKGROUND: Thoracic aortic aneurysmal diseases are characterized by degeneration of elastin within the aortic wall. Although proteinases, such as matrix metalloproteinase, appear to contribute to elastin degradation, little is known about the role of elastic fiber assembly in such diseases. Fibulin-5 is an extracellular protein that is expressed in the vascular basement membrane and regulates elastic fiber assembly by microfibril machinery. In this study, we examined whether thoracic aortic dissection (TAD) is associated with abnormal fibulin-5 expression. METHODS: Intraoperative aortic samples were obtained from 21 patients with proximal aortic dissection. Control aortic tissue was obtained from 11 organ donors, heart transplant recipients, and patients undergoing coronary artery bypass. An in vitro culture of vascular smooth muscle cells was obtained from 2 TAD patients and 1 control subject. To evaluate elastin expression, we stained tissue sections with Verhoeff-Van Gieson stain. Fibulin-5 messenger RNA (mRNA) expression was determined by quantitative real-time reverse-transcriptase-polymerase chain reaction. RESULTS: Aortic fibulin-5 mRNA and elastin content were decreased in TAD patients, compared with controls (P=.001 and P=.02, respectively). Decreased fibulin-5 expression strongly correlated with decreased amounts and fragmentation of elastin in aortic samples from patients with TAD (r=0.83, P < .0001 and F=20.7, P < .0001 respectively). The fibulin-5 mRNA in aortic vascular smooth muscle cells collected from TAD demonstrated a 38% decrease in expression, compared with the control. CONCLUSIONS: Patients with proximal aortic dissection exhibited significantly decreased expression of aortic fibulin-5. Decreased fibulin-5 may contribute to the pathogenesis of aortic dissection by impairing elastic fiber assembly.


Subject(s)
Aortic Aneurysm, Thoracic/physiopathology , Aortic Dissection/physiopathology , Elastin/genetics , Extracellular Matrix Proteins/genetics , Recombinant Proteins/genetics , Aged , Aortic Dissection/pathology , Aorta, Thoracic/pathology , Aorta, Thoracic/physiology , Aortic Aneurysm, Thoracic/pathology , Elastin/metabolism , Extracellular Matrix Proteins/metabolism , Female , Gene Expression , Humans , Male , Middle Aged , RNA, Messenger/analysis , Recombinant Proteins/metabolism
19.
mBio ; 3(4): e00159-12, 2012.
Article in English | MEDLINE | ID: mdl-22761392

ABSTRACT

UNLABELLED: Directed differentiation of stem cell lines into intestine-like tissue called induced human intestinal organoids (iHIOs) is now possible (J. R. Spence, C. N. Mayhew, S. A. Rankin, M. F. Kuhar, J. E. Vallance, K. Tolle, E. E. Hoskins, V. V. Kalinichenko, S. I. Wells, A. M. Zorn, N. F. Shroyer, and J. M. Wells, Nature 470:105-109, 2011). We tested iHIOs as a new model to cultivate and study fecal viruses. Protocols for infection of iHIOs with a laboratory strain of rotavirus, simian SA11, were developed. Proof-of-principle analyses showed that iHIOs support replication of a gastrointestinal virus, rotavirus, on the basis of detection of nonstructural viral proteins (nonstructural protein 4 [NSP4] and NSP2) by immunofluorescence, increased levels of viral RNA by quantitative reverse transcription-PCR (qRT-PCR), and production of infectious progeny virus. iHIOs were also shown to support replication of 12/13 clinical rotavirus isolates directly from stool samples. An unexpected finding was the detection of rotavirus infection not only in the epithelial cells but also in the mesenchymal cell population of the iHIOs. This work demonstrates that iHIOs offer a promising new model to study rotaviruses and other gastrointestinal viruses. IMPORTANCE: Gastrointestinal viral infections are a major cause of illness and death in children and adults. The ability to fully understand how viruses interact with human intestinal cells in order to cause disease has been hampered by insufficient methods for growing many gastrointestinal viruses in the laboratory. Induced human intestinal organoids (iHIOs) are a promising new model for generating intestine-like tissue. This is the first report of a study using iHIOs to cultivate any microorganism, in this case, an enteric virus. The evidence that both laboratory and clinical rotavirus isolates can replicate in iHIOs suggests that this model would be useful not only for studies of rotaviruses but also potentially of other infectious agents. Furthermore, detection of rotavirus proteins in unexpected cell types highlights the promise of this system to reveal new questions about pathogenesis that have not been previously recognized or investigated in other intestinal cell culture models.


Subject(s)
Intestines/virology , Organoids/virology , Rotavirus Infections/virology , Rotavirus/physiology , Stem Cells/virology , Virology/methods , Adult , Cell Differentiation , Cells, Cultured , Female , Humans , Intestines/cytology , Male , Middle Aged , Models, Biological , Organoids/cytology , Rotavirus/genetics , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL