Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Am J Respir Crit Care Med ; 207(8): 1042-1054, 2023 04 15.
Article in English | MEDLINE | ID: mdl-36480964

ABSTRACT

Rationale: Alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV) is linked to heterozygous mutations in the FOXF1 (Forkhead Box F1) gene, a key transcriptional regulator of pulmonary vascular development. There are no effective treatments for ACDMPV other than lung transplant, and new pharmacological agents activating FOXF1 signaling are urgently needed. Objectives: Identify-small molecule compounds that stimulate FOXF1 signaling. Methods: We used mass spectrometry, immunoprecipitation, and the in vitro ubiquitination assay to identify TanFe (transcellular activator of nuclear FOXF1 expression), a small-molecule compound from the nitrile group, which stabilizes the FOXF1 protein in the cell. The efficacy of TanFe was tested in mouse models of ACDMPV and acute lung injury and in human vascular organoids derived from induced pluripotent stem cells of a patient with ACDMPV. Measurements and Main Results: We identified HECTD1 as an E3 ubiquitin ligase involved in ubiquitination and degradation of the FOXF1 protein. The TanFe compound disrupted FOXF1-HECTD1 protein-protein interactions and decreased ubiquitination of the FOXF1 protein in pulmonary endothelial cells in vitro. TanFe increased protein concentrations of FOXF1 and its target genes Flk1, Flt1, and Cdh5 in LPS-injured mouse lungs, decreasing endothelial permeability and inhibiting lung inflammation. Treatment of pregnant mice with TanFe increased FOXF1 protein concentrations in lungs of Foxf1+/- embryos, stimulated neonatal lung angiogenesis, and completely prevented the mortality of Foxf1+/- mice after birth. TanFe increased angiogenesis in human vascular organoids derived from induced pluripotent stem cells of a patient with ACDMPV with FOXF1 deletion. Conclusions: TanFe is a novel activator of FOXF1, providing a new therapeutic candidate for treatment of ACDMPV and other neonatal pulmonary vascular diseases.


Subject(s)
Persistent Fetal Circulation Syndrome , Infant, Newborn , Humans , Animals , Mice , Persistent Fetal Circulation Syndrome/genetics , Endothelial Cells , Lung/metabolism , Forkhead Transcription Factors/genetics
2.
Environ Toxicol ; 38(7): 1577-1588, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36988223

ABSTRACT

Inside the biological milieu, nanoparticles with photocatalytic activity have potential to trigger cell death non-specifically due to production of reactive oxygen species (ROS) upon reacting with biological entities. Silver nanoparticle (AgNP) possessing narrow band gap energy can exhibit high light absorption property and significant photocatalytic activity. This study intends to explore the effects of ROS generated due to photocatalytic activity of AgNP on antimicrobial and cytotoxic propensities. To this end, AgNP was synthesized using the principle of green chemistry from the peel extract of Punica granatum L., and was characterized using UV-Vis spectroscope, transmission electron microscope and x-ray diffraction, and so forth. The antimicrobial activity of AgNP against studied bacteria indicated that, ROS generated at AgNP interface develop stress on bacterial membrane leading to bacterial cell death, whereas Alamar Blue dye reduction assay indicated that increased cytotoxic activity with increasing concentrations of AgNP. The γH2AX activity assay revealed that increasing the concentrations of AgNP increased DNA damaging activity. The results altogether demonstrated that both antimicrobial and cytotoxic propensities are triggered primarily due interfacial ROS generation by photocatalytic AgNP, which caused membrane deformation in bacteria and DNA damage in HT1080 cells resulting in cell death.


Subject(s)
Anti-Infective Agents , Antineoplastic Agents , Metal Nanoparticles , Reactive Oxygen Species/metabolism , Silver/toxicity , Silver/chemistry , Metal Nanoparticles/toxicity , Metal Nanoparticles/chemistry , Anti-Infective Agents/toxicity , Oxidative Stress , Antineoplastic Agents/pharmacology , Anti-Bacterial Agents/toxicity
3.
Circulation ; 144(7): 539-555, 2021 08 17.
Article in English | MEDLINE | ID: mdl-34111939

ABSTRACT

BACKGROUND: Pulmonary hypertension (PH) is a common complication in patients with alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV), a severe congenital disorder associated with mutations in the FOXF1 gene. Although the loss of alveolar microvasculature causes PH in patients with ACDMPV, it is unknown whether increasing neonatal lung angiogenesis could prevent PH and right ventricular (RV) hypertrophy. METHODS: We used echocardiography, RV catheterization, immunostaining, and biochemical methods to examine lung and heart remodeling and RV output in Foxf1WT/S52F mice carrying the S52F Foxf1 mutation (identified in patients with ACDMPV). The ability of Foxf1WT/S52F mutant embryonic stem cells to differentiate into respiratory cell lineages in vivo was examined using blastocyst complementation. Intravascular delivery of nanoparticles with a nonintegrating Stat3 expression vector was used to improve neonatal pulmonary angiogenesis in Foxf1WT/S52F mice and determine its effects on PH and RV hypertrophy. RESULTS: Foxf1WT/S52F mice developed PH and RV hypertrophy after birth. The severity of PH in Foxf1WT/S52F mice directly correlated with mortality, low body weight, pulmonary artery muscularization, and increased collagen deposition in the lung tissue. Increased fibrotic remodeling was found in human ACDMPV lungs. Mouse embryonic stem cells carrying the S52F Foxf1 mutation were used to produce chimeras through blastocyst complementation and to demonstrate that Foxf1WT/S52F embryonic stem cells have a propensity to differentiate into pulmonary myofibroblasts. Intravascular delivery of nanoparticles carrying Stat3 cDNA protected Foxf1WT/S52F mice from RV hypertrophy and PH, improved survival, and decreased fibrotic lung remodeling. CONCLUSIONS: Nanoparticle therapies increasing neonatal pulmonary angiogenesis may be considered to prevent PH in ACDMPV.


Subject(s)
Gene Transfer Techniques , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/therapy , Nanoparticles , Persistent Fetal Circulation Syndrome/complications , Pulmonary Alveoli/abnormalities , STAT3 Transcription Factor/genetics , Airway Remodeling/genetics , Animals , Biomarkers , Disease Models, Animal , Disease Susceptibility , Drug Carriers , Drug Delivery Systems , Echocardiography , Fibrosis , Forkhead Transcription Factors/deficiency , Genetic Therapy , Humans , Hypertension, Pulmonary/diagnosis , Hypertension, Pulmonary/metabolism , Hypertrophy, Right Ventricular/diagnosis , Hypertrophy, Right Ventricular/etiology , Hypertrophy, Right Ventricular/metabolism , Mice , Mice, Transgenic , Microvascular Density/genetics , Myofibroblasts/metabolism , Persistent Fetal Circulation Syndrome/genetics , Persistent Fetal Circulation Syndrome/pathology , STAT3 Transcription Factor/administration & dosage , Theranostic Nanomedicine/methods , Treatment Outcome , Vascular Remodeling/genetics
4.
Am J Respir Crit Care Med ; 202(1): 100-111, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32240596

ABSTRACT

Rationale: Advances in neonatal critical care have greatly improved the survival of preterm infants, but the long-term complications of prematurity, including bronchopulmonary dysplasia (BPD), cause mortality and morbidity later in life. Although VEGF (vascular endothelial growth factor) improves lung structure and function in rodent BPD models, severe side effects of VEGF therapy prevent its use in patients with BPD.Objectives: To test whether nanoparticle delivery of proangiogenic transcription factor FOXM1 (forkhead box M1) or FOXF1 (forkhead box F1), both downstream targets of VEGF, can improve lung structure and function after neonatal hyperoxic injury.Methods: Newborn mice were exposed to 75% O2 for the first 7 days of life before being returned to a room air environment. On Postnatal Day 2, polyethylenimine-(5) myristic acid/polyethylene glycol-oleic acid/cholesterol nanoparticles containing nonintegrating expression plasmids with Foxm1 or Foxf1 cDNAs were injected intravenously. The effects of the nanoparticles on lung structure and function were evaluated using confocal microscopy, flow cytometry, and the flexiVent small-animal ventilator.Measurements and Main Results: The nanoparticles efficiently targeted endothelial cells and myofibroblasts in the alveolar region. Nanoparticle delivery of either FOXM1 or FOXF1 did not protect endothelial cells from apoptosis caused by hyperoxia but increased endothelial proliferation and lung angiogenesis after the injury. FOXM1 and FOXF1 improved elastin fiber organization, decreased alveolar simplification, and preserved lung function in mice reaching adulthood.Conclusions: Nanoparticle delivery of FOXM1 or FOXF1 stimulates lung angiogenesis and alveolarization during recovery from neonatal hyperoxic injury. Delivery of proangiogenic transcription factors has promise as a therapy for BPD in preterm infants.


Subject(s)
Angiogenesis Inducing Agents/administration & dosage , Drug Delivery Systems , Forkhead Box Protein M1/administration & dosage , Forkhead Transcription Factors/administration & dosage , Hyperoxia/drug therapy , Nanoparticles , Pulmonary Alveoli/drug effects , Angiogenesis Inducing Agents/pharmacology , Angiogenesis Inducing Agents/therapeutic use , Animals , Animals, Newborn , Blotting, Western , Female , Flow Cytometry , Forkhead Box Protein M1/pharmacology , Forkhead Box Protein M1/therapeutic use , Forkhead Transcription Factors/pharmacology , Forkhead Transcription Factors/therapeutic use , Hyperoxia/pathology , Hyperoxia/physiopathology , Injections, Intravenous , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Pulmonary Alveoli/blood supply , Pulmonary Alveoli/pathology , Pulmonary Alveoli/physiopathology , Reverse Transcriptase Polymerase Chain Reaction , Treatment Outcome
5.
Am J Respir Crit Care Med ; 200(8): 1045-1056, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31199666

ABSTRACT

Rationale: Alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV) is a lethal congenital disorder causing respiratory failure and pulmonary hypertension shortly after birth. There are no effective treatments for ACDMPV other than lung transplant, and new therapeutic approaches are urgently needed. Although ACDMPV is linked to mutations in the FOXF1 gene, molecular mechanisms through which FOXF1 mutations cause ACDMPV are unknown.Objectives: To identify molecular mechanisms by which S52F FOXF1 mutations cause ACDMPV.Methods: We generated a clinically relevant mouse model of ACDMPV by introducing the S52F FOXF1 mutation into the mouse Foxf1 gene locus using CRISPR/Cas9 technology. Immunohistochemistry, whole-lung imaging, and biochemical methods were used to examine vasculature in Foxf1WT/S52F lungs and identify molecular mechanisms regulated by FOXF1.Measurements and Main Results: FOXF1 mutations were identified in 28 subjects with ACDMPV. Foxf1WT/S52F knock-in mice recapitulated histopathologic findings in ACDMPV infants. The S52F FOXF1 mutation disrupted STAT3-FOXF1 protein-protein interactions and inhibited transcription of Stat3, a critical transcriptional regulator of angiogenesis. STAT3 signaling and endothelial proliferation were reduced in Foxf1WT/S52F mice and human ACDMPV lungs. S52F FOXF1 mutant protein did not bind chromatin and was transcriptionally inactive. Furthermore, we have developed a novel formulation of highly efficient nanoparticles and demonstrated that nanoparticle delivery of STAT3 cDNA into the neonatal circulation restored endothelial proliferation and stimulated lung angiogenesis in Foxf1WT/S52F mice.Conclusions: FOXF1 acts through STAT3 to stimulate neonatal lung angiogenesis. Nanoparticle delivery of STAT3 is a promising strategy to treat ACDMPV associated with decreased STAT3 signaling.


Subject(s)
Forkhead Transcription Factors/genetics , Gene Expression Regulation , Mutation , Persistent Fetal Circulation Syndrome/genetics , Persistent Fetal Circulation Syndrome/physiopathology , Pulmonary Alveoli/abnormalities , Signal Transduction/genetics , Animals , Humans , Mice , Models, Animal , Pulmonary Alveoli/physiopathology
6.
J Biol Chem ; 290(12): 7563-75, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25631042

ABSTRACT

Alterations in the forkhead box F2 gene expression have been reported in numerous pathologies, and Foxf2(-/-) mice are perinatal lethal with multiple malformations; however, molecular mechanisms pertaining to Foxf2 signaling are severely lacking. In this study, Foxf2 requirements in murine smooth muscle cells were examined using a conditional knock-out approach. We generated novel Foxf2-floxed mice, which we bred to smMHC-Cre-eGFP mice to generate a mouse line with Foxf2 deleted specifically from smooth muscle. These mice exhibited growth retardation due to reduced intestinal length as well as inflammation and remodeling of the small intestine. Colons of Tg(smMHC-Cre-eGFP(+/-));Foxf2(-/-) mice had expansion of the myenteric nerve plexus and increased proliferation of smooth muscle cells leading to thickening of the longitudinal smooth muscle layer. Foxf2 deficiency in colonic smooth muscle was associated with increased expression of Foxf1, PDGFa, PDGFb, PDGF receptor α, and myocardin. FOXF2 bound to promoter regions of these genes indicating direct transcriptional regulation. Foxf2 repressed Foxf1 promoter activity in co-transfection experiments. We also show that knockdown of Foxf2 in colonic smooth muscle cells in vitro and in transgenic mice increased myocardin/serum response factor signaling and increased expression of contractile proteins. Foxf2 attenuated myocardin/serum response factor signaling in smooth muscle cells through direct binding to the N-terminal region of myocardin. Our results indicate that Foxf2 signaling in smooth muscle cells is essential for intestinal development and serum response factor signaling.


Subject(s)
Forkhead Transcription Factors/physiology , Intestines/embryology , Nuclear Proteins/metabolism , Platelet-Derived Growth Factor/metabolism , Serum Response Factor/metabolism , Signal Transduction/physiology , Trans-Activators/metabolism , Animals , Base Sequence , Chromatin Immunoprecipitation , DNA Primers , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Morphogenesis , Muscle, Smooth/metabolism , Platelet-Derived Growth Factor/genetics , Promoter Regions, Genetic
7.
Circ Res ; 115(8): 709-20, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25091710

ABSTRACT

RATIONALE: Inactivating mutations in the Forkhead Box transcription factor F1 (FOXF1) gene locus are frequently found in patients with alveolar capillary dysplasia with misalignment of pulmonary veins, a lethal congenital disorder, which is characterized by severe abnormalities in the respiratory, cardiovascular, and gastrointestinal systems. In mice, haploinsufficiency of the Foxf1 gene causes alveolar capillary dysplasia and developmental defects in lung, intestinal, and gall bladder morphogenesis. OBJECTIVE: Although FOXF1 is expressed in multiple mesenchyme-derived cell types, cellular origins and molecular mechanisms of developmental abnormalities in FOXF1-deficient mice and patients with alveolar capillary dysplasia with misalignment of pulmonary veins remain uncharacterized because of lack of mouse models with cell-restricted inactivation of the Foxf1 gene. In the present study, the role of FOXF1 in endothelial cells was examined using a conditional knockout approach. METHODS AND RESULTS: A novel mouse line harboring Foxf1-floxed alleles was generated by homologous recombination. Tie2-Cre and Pdgfb-CreER transgenes were used to delete Foxf1 from endothelial cells. FOXF1-deficient embryos exhibited embryonic lethality, growth retardation, polyhydramnios, cardiac ventricular hypoplasia, and vascular abnormalities in the lung, placenta, yolk sac, and retina. Deletion of FOXF1 from endothelial cells reduced endothelial proliferation, increased apoptosis, inhibited vascular endothelial growth factor signaling, and decreased expression of endothelial genes critical for vascular development, including vascular endothelial growth factor receptors Flt1 and Flk1, Pdgfb, Pecam1, CD34, integrin ß3, ephrin B2, Tie2, and the noncoding RNA Fendrr. Chromatin immunoprecipitation assay demonstrated that Flt1, Flk1, Pdgfb, Pecam1, and Tie2 genes are direct transcriptional targets of FOXF1. CONCLUSIONS: FOXF1 is required for the formation of embryonic vasculature by regulating endothelial genes critical for vascular development and vascular endothelial growth factor signaling.


Subject(s)
Blood Vessels/metabolism , Endothelial Cells/metabolism , Forkhead Transcription Factors/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Animals , Apoptosis/genetics , Base Sequence , Blood Vessels/embryology , Blotting, Western , Cell Line , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Developmental , Humans , Immunohistochemistry , Lung/blood supply , Lung/embryology , Lung/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid
8.
J Biol Chem ; 288(47): 33500-33508, 2013 Nov 22.
Article in English | MEDLINE | ID: mdl-24108125

ABSTRACT

Genomic instability and a predisposition to cancer are hallmarks of Bloom syndrome, an autosomal recessive disease arising from mutations in the BLM gene. BLM is a RecQ helicase component of the BLM-Topo III α-RMI1-RMI2 (BTR) complex, which maintains chromosome stability at the spindle assembly checkpoint (SAC). Other members of the BTR complex include Topo IIIa, RMI1, and RMI2. All members of the BTR complex are essential for maintaining the stable genome. Interestingly, the BTR complex is posttranslationally modified upon SAC activation during mitosis, but its significance remains unknown. In this study, we show that two proteins that interact with BLM, RMI1 and RMI2, are phosphorylated upon SAC activation, and, like BLM, RMI1, and RMI2, are phosphorylated in an MPS1-dependent manner. An S112A mutant of RMI2 localized normally in cells and was found in SAC-induced coimmunoprecipitations of the BTR complex. However, in RMI2-depleted cells, an S112A mutant disrupted the mitotic arrest upon SAC activation. The failure of cells to maintain mitotic arrest, due to lack of phosphorylation at Ser-112, results in high genomic instability characterized by micronuclei, multiple nuclei, and a wide distribution of aberrantly segregating chromosomes. We found that the S112A mutant of RMI2 showed defects in redistribution between the nucleoplasm and nuclear matrix. The phosphorylation at Ser-112 of RMI2 is independent of BLM and is not required for the stability of the BTR complex, BLM focus formation, and chromatin targeting in response to replication stress. Overall, this study suggests that the phosphorylation of the BTR complex is essential to maintain a stable genome.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Chromosomal Instability/physiology , DNA Topoisomerases, Type I/metabolism , DNA-Binding Proteins/metabolism , M Phase Cell Cycle Checkpoints/physiology , Multiprotein Complexes/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , RecQ Helicases/metabolism , Amino Acid Substitution , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , DNA Topoisomerases, Type I/genetics , DNA-Binding Proteins/genetics , HEK293 Cells , HeLa Cells , Humans , Multiprotein Complexes/genetics , Mutation, Missense , Nuclear Proteins/genetics , Phosphorylation/physiology , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , RecQ Helicases/genetics , Serine/genetics , Serine/metabolism , Spindle Apparatus/genetics , Spindle Apparatus/metabolism
9.
Blood ; 119(14): 3285-94, 2012 Apr 05.
Article in English | MEDLINE | ID: mdl-22343915

ABSTRACT

Fanconi anemia (FA) nuclear core complex is a multiprotein complex required for the functional integrity of the FA-BRCA pathway regulating DNA repair. This pathway is inactivated in FA, a devastating genetic disease, which leads to hematologic defects and cancer in patients. Here we report the isolation and characterization of a novel 20-kDa FANCA-associated protein (FAAP20). We show that FAAP20 is an integral component of the FA nuclear core complex. We identify a region on FANCA that physically interacts with FAAP20, and show that FANCA regulates stability of this protein. FAAP20 contains a conserved ubiquitin-binding zinc-finger domain (UBZ), and binds K-63-linked ubiquitin chains in vitro. The FAAP20-UBZ domain is not required for interaction with FANCA, but is required for DNA-damage-induced chromatin loading of FANCA and the functional integrity of the FA pathway. These findings reveal critical roles for FAAP20 in the FA-BRCA pathway of DNA damage repair and genome maintenance.


Subject(s)
DNA Repair , Fanconi Anemia Complementation Group Proteins/metabolism , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Signal Transduction , Ubiquitin/metabolism , Cells, Cultured , Chromatin/metabolism , DNA Damage , Fanconi Anemia Complementation Group A Protein/chemistry , Fanconi Anemia Complementation Group A Protein/metabolism , Fanconi Anemia Complementation Group Proteins/chemistry , Fanconi Anemia Complementation Group Proteins/genetics , Humans , Protein Binding , Protein Interaction Domains and Motifs , Protein Stability , Zinc Fingers
10.
Bioprocess Biosyst Eng ; 37(2): 139-49, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23719930

ABSTRACT

A biosurfactant producing microbe isolated from a river bank was identified as Lysinibacillus fusiformis S9. It was identified with help of biochemical tests and 16S rRNA gene phylogenetic analysis. The biosurfactant S9BS produced was purified and characterized as glycolipid. The biosurfactant showed remarkable inhibition of biofilm formation by pathogenic bacteria like Escherichia coli and Streptococcus mutans. It was interesting to note that at concentration of 40 µg ml(-1) the biosurfactant did not show any bactericidal activity but restricted the biofilm formation completely. L. fusiformis is reported for the first time to produce a glycolipid type of biosurfactant capable of inhibiting biofilm formation by pathogenic bacteria. The biosurfactant inhibited bacterial attachment and biofilm formation equally well on hydrophilic as well as hydrophobic surfaces like glass and catheter tubing. This property is significant in many biomedical applications where the molecule should help in preventing biofouling of surfaces without being toxic to biotic system.


Subject(s)
Bacillaceae/metabolism , Biofilms/drug effects , Surface-Active Agents/metabolism , Bacillaceae/classification , Bacillaceae/genetics , Base Sequence , Culture Media , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Phylogeny , RNA, Ribosomal, 16S/genetics , Spectroscopy, Fourier Transform Infrared , Surface-Active Agents/pharmacology
11.
Hum Gene Ther ; 35(1-2): 59-69, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38062776

ABSTRACT

Despite decades of research in adeno-associated virus (AAV) and the role of adenovirus in production, the interplay of AAV and adenovirus is not fully understood. Specific regions of the adenoviral genome containing E1, E2a, E4 open reading frame (ORF), and VA RNA have been demonstrated as necessary for AAV production; however, incorporating these regions into either a producer cell line or subcloning into an Ad helper plasmid may lead to inclusion of neighboring adenoviral sequence or ORFs with unknown function. Because AAV is frequently used in gene therapies, removing excessive adenovirus sequences improves the Ad helper plasmid size and manufacturability, and may lead to safer vectors for patients. Furthermore, deepening our understanding of the helper virus genes required for recombinant AAV (rAAV) production has the potential to increase yields and manufacturability of rAAV for clinical and commercial applications. One region continuously included in various Ad helper plasmid iterations is the adenoviral E2a promoter region that appears to be necessary for E2a expression. Due to the compact nature of viral genomes, the E2a promoter region overlaps with the Hexon Assembly/100K protein and the L4 region. The L4 region, which contains the coding sequences for 22K and 33K proteins, had not been thought to be necessary for AAV production. Through molecular techniques, this study demonstrates that the adenoviral 22K protein is essential for rAAV production in HEK293 cells by triple transfection and that the 33K protein synergistically increases rAAV yield.


Subject(s)
Adenoviridae , Dependovirus , Humans , Dependovirus/genetics , Dependovirus/metabolism , Adenoviridae/genetics , HEK293 Cells , Plasmids , Transfection , Viral Proteins/genetics , Genetic Vectors/genetics
12.
Int J Biol Macromol ; 261(Pt 2): 129848, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38302032

ABSTRACT

Glycolipid-based biosurfactants (BSs), known for their intriguing and diverse properties, represent a largely uncharted territory in the realm of potential biomedical applications. This field holds great promise yet remains largely unexplored. This investigation provides new insights into the isolation, characterization, and comprehensive biomedical assessment of a novel glycolipid biosurfactant derived from Bacillus species, meeting the growing demand for understanding its multifaceted impact on various biomedical issues. Within this framework, two glycolipids, BG2A and BG2B, emerged as the most proficient strains in biosurfactant (BS) production. The biosurfactants (BSs) ascertained as glycolipids via thin layer chromatography (TLC) exhibited antimicrobial activity against S. aureus and E. coli. Both isolates exhibited anticancer effects against cervical carcinoma cells and demonstrated significant anti-biofilm activity against V. cholerae. Moreover, molecular docking and molecular dynamics (MD) simulations were employed to explore their antimicrobial resistance properties against Tyrosyl-tRNA synthetase (TyrRS) of Staphylococcus aureus, a well-annotated molecular target. Characterization and interpretation using Fourier transform infrared spectroscopy (FTIR) and nuclear magnetic resonance spectroscopy (1H and 13C NMR) confirmed that the BSs produced by each strain were glycolipids. These findings suggest that the isolated BSs can serve as effective agents with antibiofilm, antimicrobial, antioxidant, and anticancer properties, in addition to their considerable antibacterial resistance attributes.


Subject(s)
Anti-Infective Agents , Bacillus , Tyrosine-tRNA Ligase , Staphylococcus aureus , Molecular Docking Simulation , Molecular Dynamics Simulation , Glycolipids/pharmacology , Glycolipids/chemistry , Escherichia coli , Surface-Active Agents/chemistry , Anti-Infective Agents/pharmacology
13.
Sci Rep ; 12(1): 8383, 2022 05 19.
Article in English | MEDLINE | ID: mdl-35589849

ABSTRACT

The green synthesis of silver nanoparticles (AgNPs) and their applications have attracted many researchers as the AgNPs are used effectively in targeting specific tissues and pathogenic microorganisms. The purpose of this study is to synthesize and characterize silver nanoparticles from fully expanded leaves of Eugenia roxburghii DC., as well as to test their effectiveness in inhibiting biofilm production. In this study, at 0.1 mM concentration of silver nitrate (AgNO3), stable AgNPs were synthesized and authenticated by monitoring the color change of the solution from yellow to brown, which was confirmed with spectrophotometric detection of optical density. The crystalline nature of these AgNPs was detected through an X-Ray Diffraction (XRD) pattern. AgNPs were characterized through a high-resolution transmission electron microscope (HR-TEM) to study the morphology and size of the nanoparticles (NPs). A new biological approach was undertaken through the Congo Red Agar (CRA) plate assay by using the synthesized AgNPs against biofilm production. The AgNPs effectively inhibit biofilm formation and the biofilm-producing bacterial colonies. This could be a significant achievement in contending with many dynamic pathogens.


Subject(s)
Eugenia , Metal Nanoparticles , Anti-Bacterial Agents/chemistry , Bacteria , Biofilms , Green Chemistry Technology , Metal Nanoparticles/chemistry , Microbial Sensitivity Tests , Plant Extracts/chemistry , Plant Extracts/pharmacology , Silver/pharmacology , X-Ray Diffraction
14.
Nat Commun ; 13(1): 2080, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35440116

ABSTRACT

Pulmonary endothelial progenitor cells (EPCs) are critical for neonatal lung angiogenesis and represent a subset of general capillary cells (gCAPs). Molecular mechanisms through which EPCs stimulate lung angiogenesis are unknown. Herein, we used single-cell RNA sequencing to identify the BMP9/ACVRL1/SMAD1 pathway signature in pulmonary EPCs. BMP9 receptor, ACVRL1, and its downstream target genes were inhibited in EPCs from Foxf1WT/S52F mutant mice, a model of alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV). Expression of ACVRL1 and its targets were reduced in lungs of ACDMPV subjects. Inhibition of FOXF1 transcription factor reduced BMP9/ACVRL1 signaling and decreased angiogenesis in vitro. FOXF1 synergized with ETS transcription factor FLI1 to activate ACVRL1 promoter. Nanoparticle-mediated silencing of ACVRL1 in newborn mice decreased neonatal lung angiogenesis and alveolarization. Treatment with BMP9 restored lung angiogenesis and alveolarization in ACVRL1-deficient and Foxf1WT/S52F mice. Altogether, EPCs promote neonatal lung angiogenesis and alveolarization through FOXF1-mediated activation of BMP9/ACVRL1 signaling.


Subject(s)
Endothelial Progenitor Cells , Persistent Fetal Circulation Syndrome , Pneumonia , Animals , Mice , Activin Receptors, Type II/metabolism , Endothelial Progenitor Cells/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Lung/metabolism , Persistent Fetal Circulation Syndrome/genetics , Persistent Fetal Circulation Syndrome/metabolism , Pneumonia/metabolism , Pulmonary Alveoli/abnormalities
15.
Oncogene ; 40(12): 2182-2199, 2021 03.
Article in English | MEDLINE | ID: mdl-33627785

ABSTRACT

The PAX3-FOXO1 fusion protein is the key oncogenic driver in fusion positive rhabdomyosarcoma (FP-RMS), an aggressive soft tissue malignancy with a particularly poor prognosis. Identifying key downstream targets of PAX3-FOXO1 will provide new therapeutic opportunities for treatment of FP-RMS. Herein, we demonstrate that Forkhead Box F1 (FOXF1) transcription factor is uniquely expressed in FP-RMS and is required for FP-RMS tumorigenesis. The PAX3-FOXO1 directly binds to FOXF1 enhancers and induces FOXF1 gene expression. CRISPR/Cas9 mediated inactivation of either FOXF1 coding sequence or FOXF1 enhancers suppresses FP-RMS tumorigenesis even in the presence of PAX3-FOXO1 oncogene. Knockdown or genetic knockout of FOXF1 induces myogenic differentiation in PAX3-FOXO1-positive FP-RMS. Over-expression of FOXF1 decreases myogenic differentiation in primary human myoblasts. In FP-RMS tumor cells, FOXF1 protein binds chromatin near enhancers associated with FP-RMS gene signature. FOXF1 cooperates with PAX3-FOXO1 and E-box transcription factors MYOD1 and MYOG to regulate FP-RMS-specific gene expression. Altogether, FOXF1 functions downstream of PAX3-FOXO1 to promote FP-RMS tumorigenesis.


Subject(s)
Carcinogenesis/genetics , Forkhead Box Protein O1/genetics , Forkhead Transcription Factors/genetics , PAX3 Transcription Factor/genetics , Rhabdomyosarcoma/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , Humans , Muscle Development/genetics , MyoD Protein/genetics , Myogenin/genetics , Rhabdomyosarcoma/pathology
16.
Curr Stem Cell Rep ; 6(3): 41-51, 2020 Sep.
Article in English | MEDLINE | ID: mdl-33184603

ABSTRACT

PURPOSE OF THE REVIEW: Significant numbers of patients worldwide are affected by various rare diseases, but the effective treatment options to these individuals are limited. Rare diseases remain underfunded compared to more common diseases, leading to significant delays in research progress and ultimately, to finding an effective cure. Here, we review the use of genome-editing tools to understand the pathogenesis of rare diseases and develop additional therapeutic approaches with a high degree of precision. RECENT FINDINGS: Several genome-editing approaches, including CRISPR/Cas9, TALEN and ZFN, have been used to generate animal models of rare diseases, understand the disease pathogenesis, correct pathogenic mutations in patient-derived somatic cells and iPSCs, and develop new therapies for rare diseases. The CRISPR/Cas9 system stands out as the most extensively used method for genome editing due to its relative simplicity and superior efficiency compared to TALEN and ZFN. CRISPR/Cas9 is emerging as a feasible gene-editing option to treat rare monogenic and other genetically defined human diseases. SUMMARY: Less than 5% of ~7000 known rare diseases have FDA-approved therapies, providing a compelling need for additional research and clinical trials to identify efficient treatment options for patients with rare diseases. Development of efficient genome-editing tools capable to correct or replace dysfunctional genes will lead to novel therapeutic approaches in these diseases.

17.
Mol Biol Cell ; 31(13): 1411-1424, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32348194

ABSTRACT

Forkhead box M1 (FOXM1), a nuclear transcription factor that activates cell cycle regulatory genes, is highly expressed in a majority of human cancers. The function of FOXM1 independent of nuclear transcription is unknown. In the present study, we found the FOXM1 protein inside the mitochondria. Using site-directed mutagenesis, we generated FOXM1 mutant proteins that localized to distinct cellular compartments, uncoupling the nuclear and mitochondrial functions of FOXM1. Directing FOXM1 into the mitochondria decreased mitochondrial mass, membrane potential, respiration, and electron transport chain (ETC) activity. In mitochondria, the FOXM1 directly bound to and increased the pentatricopeptide repeat domain 1 (PTCD1) protein, a mitochondrial leucine-specific tRNA binding protein that inhibits leucine-rich ETC complexes. Mitochondrial FOXM1 did not change cellular proliferation. Thus, FOXM1 translocates into mitochondria and inhibits mitochondrial respiration by increasing PTCD1. We identify a new paradigm that FOXM1 regulates mitochondrial homeostasis in a process independent of nuclear transcription.


Subject(s)
Forkhead Box Protein M1/metabolism , Mitochondria/metabolism , Oxidative Phosphorylation , Animals , Computer Simulation , Forkhead Box Protein M1/genetics , Gene Expression Regulation , Humans , Mice , Mitochondrial Proteins/genetics , Mutation , RNA-Binding Proteins/genetics , Rats , Xenopus laevis , Zebrafish
18.
Mol Cancer Ther ; 18(7): 1217-1229, 2019 07.
Article in English | MEDLINE | ID: mdl-31040162

ABSTRACT

The oncogenic transcription factor FOXM1 has been previously shown to play a critical role in carcinogenesis by inducing cellular proliferation in multiple cancer types. A small-molecule compound, Robert Costa Memorial drug-1 (RCM-1), has been recently identified from high-throughput screen as an inhibitor of FOXM1 in vitro and in mouse model of allergen-mediated lung inflammation. In the present study, we examined antitumor activities of RCM-1 using tumor models. Treatment with RCM-1 inhibited tumor cell proliferation as evidenced by increased cell-cycle duration. Confocal imaging of RCM-1-treated tumor cells indicated that delay in cellular proliferation was concordant with inhibition of FOXM1 nuclear localization in these cells. RCM-1 reduced the formation and growth of tumor cell colonies in the colony formation assay. In animal models, RCM-1 treatment inhibited growth of mouse rhabdomyosarcoma Rd76-9, melanoma B16-F10, and human H2122 lung adenocarcinoma. RCM-1 decreased FOXM1 protein in the tumors, reduced tumor cell proliferation, and increased tumor cell apoptosis. RCM-1 decreased protein levels and nuclear localization of ß-catenin, and inhibited protein-protein interaction between ß-catenin and FOXM1 in cultured tumor cells and in vivo Altogether, our study provides important evidence of antitumor potential of the small-molecule compound RCM-1, suggesting that RCM-1 can be a promising candidate for anticancer therapy.


Subject(s)
Forkhead Box Protein M1/genetics , beta Catenin/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Mice , Transfection , beta Catenin/genetics
19.
Gene ; 420(1): 66-75, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18571876

ABSTRACT

Helicases are essential enzymes, which play important role in the metabolism of nucleic acids. In the present study we report further characterization of PfH45 (Plasmodium falciparum helicase 45), which is an essential enzyme for parasite survival. The results show that the helicase activity of PfH45 is significantly stimulated by replication fork like structure. The studies using truncated derivatives of PfH45 show that its nucleic acid dependent ATPase activity resides in the N-terminal one third of the protein and its RNA and DNA-binding activity predominantly resides in the C-terminal two third of the protein. The phosphorylation of PfH45 by protein kinase C at Ser and Thr residues stimulated its DNA and RNA helicase and ssDNA and RNA-dependent ATPase activities. DNA-interacting compounds actinomycin, DAPI, daunorubicin, ethidium bromide, netropsin and nogalamycin were able to inhibit the helicase and ssDNA-dependent ATPase activity with apparent IC50 values ranging from 0.5 to 5.0 microM respectively. These compounds distinctively inhibit the helicase activity probably by forming complex with DNA and obstructing enzyme movement.


Subject(s)
DNA Helicases/chemistry , DNA Helicases/metabolism , Plasmodium falciparum/enzymology , Adenosine Triphosphatases/metabolism , Animals , DNA Replication , Nucleic Acid Conformation , Phosphorylation , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , RNA-Binding Proteins/metabolism
20.
Mol Biochem Parasitol ; 157(2): 137-47, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18054093

ABSTRACT

The human malaria parasite Plasmodium falciparum exports a variety of its proteins through its endoplasmic reticulum (ER) based secretory pathway in order to survive in the host erythrocyte. Signal peptidases are membrane-bound endopeptidases and have an important role in the transport and maturation of these parasite proteins. Prokaryotic signal peptidases are indispensable enzymes required for the removal of N-terminal signal peptide from the secretory proteins. Eukaryotic signal peptidases exist as multimeric protein complex in the ER and the catalytic subunit of this complex catalyzes removal of the N-terminal signal peptide from preproteins. All the signal peptidases contain five regions of high-sequence similarity referred to as boxes A-E. Here we report characterization of the catalytic subunit of signal peptidase complex (SPC) from P. falciparum. This protein designated as PfSP21 shows homology with the similar subunit from other sources and contains all the conserved boxes A-E. PfSP21 is able to cleave the peptide substrate containing the signal peptidase cleavage site. PfSP21 is phosphorylated by protein kinase C and its enzyme activity was upregulated after this phosphorylation. Immunofluorescence assay studies revealed that PfSP21 is localized in the ER of P. falciparum. PfSP21 dsRNA specifically inhibits the growth of P. falciparum in culture and this inhibition is most likely due to the decrease in the amount of endogenous PfSP21 protein. These studies demonstrate the characterization of a functional subunit of SPC from P. falciparum and should make an important contribution in our better understanding of the complex process of protein translocation in the parasite.


Subject(s)
Membrane Proteins/metabolism , Plasmodium falciparum/physiology , Protozoan Proteins/metabolism , Serine Endopeptidases/metabolism , Animals , Conserved Sequence , DNA, Protozoan/chemistry , DNA, Protozoan/genetics , Endoplasmic Reticulum/chemistry , Humans , Membrane Proteins/genetics , Microscopy, Fluorescence , Molecular Sequence Data , Peptides/genetics , Peptides/metabolism , Phosphorylation , Plasmodium falciparum/genetics , Protein Kinase C/metabolism , Protozoan Proteins/genetics , RNA Interference , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Serine Endopeptidases/genetics , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL