Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
Add more filters

Publication year range
1.
J Biol Chem ; 299(12): 105408, 2023 12.
Article in English | MEDLINE | ID: mdl-38229397

ABSTRACT

Increased endothelial cell (EC) permeability is a cardinal feature of acute lung injury/acute respiratory distress syndrome (ALI/ARDS). Tyrosine phosphorylation of VE-cadherin is a key determinant of EC barrier disruption. However, the identity and role of tyrosine kinases in this context are incompletely understood. Here we report that Spleen Tyrosine Kinase (Syk) is a key mediator of EC barrier disruption and lung vascular leak in sepsis. Inhibition of Syk by pharmacological or genetic approaches, each reduced thrombin-induced EC permeability. Mechanistically, Syk associates with and phosphorylates VE-cadherin to cause EC permeability. To study the causal role of endothelial Syk in sepsis-induced ALI, we used a remarkably efficient and cost-effective approach based on gene transfer to generate EC-ablated Syk mice. These mice were protected against sepsis-induced loss of VE-cadherin and inflammatory lung injury. Notably, the administration of Syk inhibitor R788 (fostamatinib); currently in phase II clinical trial for the treatment of COVID-19, mitigated lung injury and mortality in mice with sepsis. These data identify Syk as a novel kinase for VE-cadherin and a druggable target against ALI in sepsis.


Subject(s)
Acute Lung Injury , Antigens, CD , Cadherins , Respiratory Distress Syndrome , Sepsis , Syk Kinase , Animals , Mice , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Antigens, CD/metabolism , Cadherins/metabolism , Capillary Permeability , Lung/metabolism , Sepsis/complications , Syk Kinase/metabolism , Phosphorylation
2.
Mol Pharm ; 20(12): 6090-6103, 2023 Dec 04.
Article in English | MEDLINE | ID: mdl-37963105

ABSTRACT

Short-interfering RNA (siRNA) oligonucleotide therapeutics that modify gene expression by accessing RNA-interference (RNAi) pathways have great promise for the treatment of a range of disorders; however, their application in clinical settings has been limited by significant challenges in cellular delivery. Herein, we report a structure-function study using a series of modified cyclic amphipathic cell-penetrating peptides (CAPs) to determine the impact of peptide sequence on (1) siRNA-binding efficiency, (2) cellular delivery and knockdown efficiency, and (3) the endocytic uptake mechanism. Nine cyclic peptides of the general sequence Ac-C[XZ]4CG-NH2 in which X residues are hydrophobic/aromatic (Phe, Tyr, Trp, or Leu) and Z residues are charged/hydrophilic (Arg, Lys, Ser, or Glu) are assessed along with one acyclic peptide, Ac-(WR)4G-NH2. Cyclization is enforced by intramolecular disulfide bond formation between the flanking Cys residues. Binding analyses indicate that strong cationic character and the presence of aromatic residues that are competent to participate in CH-π interactions lead to CAP sequences that most effectively interact with siRNA. CAP-siRNA binding increases in the following order as a function of CAP hydrophobic/aromatic content: His < Phe < Tyr < Trp. Both cationic charge and disulfide-constrained cyclization of CAPs improve uptake of siRNA in vitro. Net neutral CAPs and an acyclic peptide demonstrate less-efficient siRNA translocation compared to the cyclic, cationic CAPs tested. All CAPs tested facilitated efficient siRNA target gene knockdown of at least 50% (as effective as a lipofectamine control), with the best CAPs enabling >80% knockdown. Significantly, gene knockdown efficiency does not strongly correlate with CAP-siRNA internalization efficiency but moderately correlates with CAP-siRNA-binding affinity. Finally, utilization of small-molecule inhibitors and targeted knockdown of essential endocytic pathway proteins indicate that most CAP-siRNA nanoparticles facilitate siRNA delivery through clathrin- and caveolin-mediated endocytosis. These results provide insight into the design principles for CAPs to facilitate siRNA delivery and the mechanisms by which these peptides translocate siRNA into cells. These studies also demonstrate the nature of the relationships between peptide-siRNA binding, cellular delivery of siRNA cargo, and functional gene knockdown. Strong correlations between these properties are not always observed, which illustrates the complexity in the design of optimal next-generation materials for oligonucleotide delivery.


Subject(s)
Cell-Penetrating Peptides , Peptides, Cyclic , Peptides, Cyclic/chemistry , RNA, Small Interfering/chemistry , Gene Knockdown Techniques , Cell-Penetrating Peptides/chemistry , Oligonucleotides , Disulfides
3.
J Med Genet ; 57(5): 296-300, 2020 05.
Article in English | MEDLINE | ID: mdl-31662342

ABSTRACT

BACKGROUND: Alveolar capillary dysplasia with misalignment of the pulmonary veins (ACDMPV) is a lethal disorder of lung development. ACDMPV is associated with haploinsufficiency of the transcription factor FOXF1, which plays an important role in the development of the lung and intestine. CNVs upstream of the FOXF1 gene have also been associated with an ACDMPV phenotype, but mechanism(s) by which these deletions disrupt lung development are not well understood. The objective of our study is to gain insights into the mechanisms by which CNVs contribute to an ACDMPV phenotype. METHODS: We analysed primary lung tissue from an infant with classic clinical and histological findings of ACDMPV and harboured a 340 kb deletion on chromosome 16q24.1 located 250 kb upstream of FOXF1. RESULTS: In RNA generated from paraffin-fixed lung sections, our patient had lower expression of FOXF1 than age-matched controls. He also had an abnormal pattern of FOXF1 protein expression, with a dramatic loss of FOXF1 expression in the lung. To gain insights into the mechanisms underlying these changes, we assessed the epigenetic landscape using chromatin immunoprecipitation, which demonstrated loss of histone H3 lysine 27 acetylation (H3K27Ac), an epigenetic mark of active enhancers, in the region of the deletion. CONCLUSIONS: Together, these data suggest that the deletion disrupts an enhancer responsible for directing FOXF1 expression in the developing lung and provide novel insights into the mechanisms underlying a fatal developmental lung disorder.


Subject(s)
Forkhead Transcription Factors/genetics , Genetic Predisposition to Disease , Lung/metabolism , Persistent Fetal Circulation Syndrome/genetics , Chromosomes, Human, Pair 16/genetics , Enhancer Elements, Genetic/genetics , Gene Deletion , Gene Expression Regulation/genetics , Haploinsufficiency/genetics , Humans , Infant , Infant, Newborn , Lung/growth & development , Lung/pathology , Persistent Fetal Circulation Syndrome/pathology
4.
N Engl J Med ; 376(5): 440-450, 2017 02 02.
Article in English | MEDLINE | ID: mdl-27959709

ABSTRACT

BACKGROUND: Continuous-flow left ventricular assist systems increase the rate of survival among patients with advanced heart failure but are associated with the development of pump thrombosis. We investigated the effects of a new magnetically levitated centrifugal continuous-flow pump that was engineered to avert thrombosis. METHODS: We randomly assigned patients with advanced heart failure to receive either the new centrifugal continuous-flow pump or a commercially available axial continuous-flow pump. Patients could be enrolled irrespective of the intended goal of pump support (bridge to transplantation or destination therapy). The primary end point was a composite of survival free of disabling stroke (with disabling stroke indicated by a modified Rankin score >3; scores range from 0 to 6, with higher scores indicating more severe disability) or survival free of reoperation to replace or remove the device at 6 months after implantation. The trial was powered for noninferiority testing of the primary end point (noninferiority margin, -10 percentage points). RESULTS: Of 294 patients, 152 were assigned to the centrifugal-flow pump group and 142 to the axial-flow pump group. In the intention-to-treat population, the primary end point occurred in 131 patients (86.2%) in the centrifugal-flow pump group and in 109 (76.8%) in the axial-flow pump group (absolute difference, 9.4 percentage points; 95% lower confidence boundary, -2.1 [P<0.001 for noninferiority]; hazard ratio, 0.55; 95% confidence interval [CI], 0.32 to 0.95 [two-tailed P=0.04 for superiority]). There were no significant between-group differences in the rates of death or disabling stroke, but reoperation for pump malfunction was less frequent in the centrifugal-flow pump group than in the axial-flow pump group (1 [0.7%] vs. 11 [7.7%]; hazard ratio, 0.08; 95% CI, 0.01 to 0.60; P=0.002). Suspected or confirmed pump thrombosis occurred in no patients in the centrifugal-flow pump group and in 14 patients (10.1%) in the axial-flow pump group. CONCLUSIONS: Among patients with advanced heart failure, implantation of a fully magnetically levitated centrifugal-flow pump was associated with better outcomes at 6 months than was implantation of an axial-flow pump, primarily because of the lower rate of reoperation for pump malfunction. (Funded by St. Jude Medical; MOMENTUM 3 ClinicalTrials.gov number, NCT02224755 .).


Subject(s)
Heart Failure/therapy , Heart-Assist Devices , Adult , Aged , Aged, 80 and over , Disease-Free Survival , Heart Failure/mortality , Heart-Assist Devices/adverse effects , Humans , Intention to Treat Analysis , Kaplan-Meier Estimate , Middle Aged , Prosthesis Design , Prosthesis Failure , Stroke/etiology , Thrombosis/etiology , Young Adult
5.
Biochem Biophys Res Commun ; 488(2): 253-258, 2017 06 24.
Article in English | MEDLINE | ID: mdl-28412356

ABSTRACT

CRM1 (chromosome maintenance region 1, Exportin 1) binds to nuclear export signals and is required for nucleocytoplasmic transport of a large variety of proteins and RNP complexes. Leptomycin B (LMB), the first specific inhibitor of CRM1 identified, binds covalently to cysteine 528 in the nuclear export signal binding region of CRM1 leading to the inhibition of protein nuclear export. Although the biochemical mechanisms of action of CRM1 inhibitors such as LMB are well studied, the subcellular effects of inhibition on CRM1 are unknown. We have found that LMB causes CRM1 to redistribute from the nucleus to the cytoplasm in A549 cells. A significant decrease in nuclear CRM1 coupled with an increase in cytoplasmic CRM1 was sustained for up to 4 h, while there was no change in total CRM1 protein in fractionated cells. Cells expressing an LMB insensitive HA-tagged CRM1-C528S protein were unaffected by LMB treatment, whereas HA-tagged wildtype CRM1 redistributed from the nucleus to the cytoplasm with LMB treatment, similar to endogenous CRM1. GFP-tagged CRM1 protein microinjected into the cytoplasm of A549 cells distributed throughout the cell in untreated cells remained primarily cytoplasmic in LMB-treated cells. Upon nuclear microinjection, GFP-CRM1 translocated to and accumulated in the cytoplasm of LMB-treated cells. Thus, LMB binds to CRM1 and causes its redistribution to the cytoplasm by inhibiting its nuclear import. Decreasing the nuclear availability of CRM1 likely contributes to the accumulation of CRM1 cargo proteins in the nucleus, suggesting a new mechanism of action for LMB.


Subject(s)
Karyopherins/metabolism , Protein Transport/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , Active Transport, Cell Nucleus/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytoplasm/drug effects , Cytoplasm/metabolism , Dose-Response Relationship, Drug , Fatty Acids, Unsaturated/pharmacology , Humans , Karyopherins/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Structure-Activity Relationship , Tumor Cells, Cultured , Exportin 1 Protein
6.
J Card Surg ; 30(10): 775-80, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26283153

ABSTRACT

BACKGROUND: Recent reports suggested that HeartMate II (HMII) thrombosis rates may be higher in implants after 2011. We characterize events at HMII centers (>100 HMII implants) whose device thrombosis rates are equivalent or lower than reported by INTERMACS. METHODS: Seven centers pooled implants from 2011 through June 2013 to examine pump thrombus and identify characteristics and clinical strategies that potentially mitigate the risk. A total of 666 patients (age 59 ± 13 years; 81% male) were studied (support duration: 13.7 ± 8.3 months, cumulative: 759 patient years). Median target INR was 2.25 (range 2.0 to 2.5), and median pump speed was 9200 rpm (range 8600 to 9600). Pump thrombus was suspected with clinical evidence (e.g., hemolysis, positive ramp test) requiring intervention (e.g., anticoagulation therapy, pump exchange) or patient death. RESULTS: Suspected pump thrombus occurred in 24/666 (3.6%) patients within three months of implant. At six months, 38/666 (5.7%) had suspected pump thrombus including 24 (3.6%) resulting in pump exchange or death. Stroke (hemorrhagic: 0.049, and ischemic: 0.048 events/patient year) and survival (six months: 88 ± 1%; 1 year: 81 ± 2%) were consistent with national averages. Suspected pump thrombus patients were younger (55 ± 13 vs. 59 ± 13, p = 0.046) and had more females (31.6% vs. 18.3%, p = 0.054). There was no difference in indication, etiology of heart failure, or body size. CONCLUSIONS: This analysis demonstrates low HMII thrombus events. Minimization of risk factors by uniform implant techniques and consistent post-op management may reduce device thrombosis. A larger scale multicenter evaluation may better elucidate the difference in thrombus events between centers.


Subject(s)
Heart Ventricles , Heart-Assist Devices/adverse effects , Thrombosis/epidemiology , Thrombosis/etiology , Adult , Age Factors , Aged , Female , Humans , Male , Middle Aged , Prosthesis Implantation/methods , Risk Factors , Sex Factors , Stroke/epidemiology , Stroke/etiology , Stroke/prevention & control , Thrombosis/prevention & control , Time Factors
7.
Int J Mol Sci ; 16(8): 19369-86, 2015 Aug 17.
Article in English | MEDLINE | ID: mdl-26287182

ABSTRACT

Efficient non-viral plasmid DNA transfection of most stem cells, progenitor cells and primary cell lines currently presents an obstacle for many applications within gene therapy research. From a standpoint of efficiency and cell viability, magnetic nanoparticle-based DNA transfection is a promising gene vectoring technique because it has demonstrated rapid and improved transfection outcomes when compared to alternative non-viral methods. Recently, our research group introduced oscillating magnet arrays that resulted in further improvements to this novel plasmid DNA (pDNA) vectoring technology. Continued improvements to nanomagnetic transfection techniques have focused primarily on magnetic nanoparticle (MNP) functionalization and transfection parameter optimization: cell confluence, growth media, serum starvation, magnet oscillation parameters, etc. Noting that none of these parameters can assist in the nuclear translocation of delivered pDNA following MNP-pDNA complex dissociation in the cell's cytoplasm, inclusion of a cassette feature for pDNA nuclear translocation is theoretically justified. In this study incorporation of a DNA targeting sequence (DTS) feature in the transfecting plasmid improved transfection efficiency in model neurons, presumably from increased nuclear translocation. This observation became most apparent when comparing the response of the dividing SH-SY5Y precursor cell to the non-dividing and differentiated SH-SY5Y neuroblastoma cells.


Subject(s)
DNA/administration & dosage , Magnetite Nanoparticles , Neurons/metabolism , Plasmids/administration & dosage , Transfection/methods , Base Sequence , Cell Line , DNA/genetics , Humans , Magnetics/methods , Magnetite Nanoparticles/chemistry , Plasmids/genetics
8.
Mol Ther ; 21(4): 775-85, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23164933

ABSTRACT

One of the barriers to successful nonviral gene delivery is the crowded cytoplasm, which plasmids need to actively traverse for gene expression. Relatively little is known about how this process occurs, but our lab and others have shown that the microtubule network and motors are required for plasmid movement to the nucleus. To further investigate how plasmids exploit normal physiological processes to transfect cells, we have taken a proteomics approach to identify the proteins that comprise the plasmid-trafficking complex. We have developed a live cell DNA-protein pull-down assay to isolate complexes at certain time points post-transfection (15 minutes to 4 hours) for analysis by mass spectrometry (MS). Plasmids containing promoter sequences bound hundreds of unique proteins as early as 15 minutes post-electroporation, while a plasmid lacking any eukaryotic sequences failed to bind many of the proteins. Specific proteins included microtubule-based motor proteins (e.g., kinesin and dynein), proteins involved in protein nuclear import (e.g., importin 1, 2, 4, and 7, Crm1, RAN, and several RAN-binding proteins), a number of heterogeneous nuclear ribonucleoprotein (hnRNP)- and mRNA-binding proteins, and transcription factors. The significance of several of the proteins involved in protein nuclear localization and plasmid trafficking was determined by monitoring movement of microinjected fluorescently labeled plasmids via live cell particle tracking in cells following protein knockdown by small-interfering RNA (siRNA) or through the use of specific inhibitors. While importin ß1 was required for plasmid trafficking and subsequent nuclear import, importin α1 played no role in microtubule trafficking but was required for optimal plasmid nuclear import. Surprisingly, the nuclear export protein Crm1 also was found to complex with the transfected plasmids and was necessary for plasmid trafficking along microtubules and nuclear import. Our results show that various proteins involved in nuclear import and export influence intracellular trafficking of plasmids and subsequent nuclear accumulation.


Subject(s)
DNA/metabolism , Proteins/metabolism , Proteomics/methods , Blotting, Western , Cell Line , Cell Line, Tumor , Gene Transfer Techniques , Humans , Mass Spectrometry , Plasmids/genetics , Protein Binding , Tandem Mass Spectrometry
9.
J Membr Biol ; 246(10): 737-44, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23525583

ABSTRACT

The use of electroporation to facilitate gene transfer is an extremely powerful and useful method for both in vitro and in vivo applications. One of its great strengths is that it induces functional destabilization and permeabilization of cell membranes throughout a tissue leading to widespread gene transfer to multiple cells and cell types within the electric field. While this is a strength, it can also be a limitation in terms of cell-specific gene delivery. The ability to restrict gene delivery and expression to particular cell types is of paramount importance for many types of gene therapy, since ectopic expression of a transgene could lead to deleterious host inflammatory responses or dysregulation of normal cellular functions. At present, there are relatively few ways to obtain cell-specific targeting of nonviral vectors, molecular probes, small molecules, and imaging agents. We have developed a novel means of restricting gene delivery to desired cell types based on the ability to control the transport of plasmids into the nuclei of desired cell types. In this article, we discuss the mechanisms of this approach and several applications in living animals to demonstrate the benefits of the combination of electroporation and selective nuclear import of plasmids for cell-specific gene delivery.


Subject(s)
Electroporation , Gene Transfer Techniques , Animals , Biological Transport , Cell Nucleus/metabolism , DNA/genetics , DNA/metabolism , Electroporation/methods , Gene Expression , Genetic Therapy , Humans , Plasmids/genetics , Plasmids/metabolism , Transgenes
10.
Am J Pathol ; 181(2): 441-51, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22677423

ABSTRACT

Oxygen exposure in premature infants is a major risk factor for bronchopulmonary dysplasia and can impair the host response to respiratory viral infections later in life. Similarly, adult mice exposed to hyperoxia as neonates display alveolar simplification associated with a reduced number of alveolar epithelial type II cells and exhibit persistent inflammation, fibrosis, and mortality when infected with influenza A virus. Because type II cells participate in innate immunity and alveolar repair, their loss may contribute to oxygen-mediated sensitivity to viral infection. A genomewide screening of type II cells identified eosinophil-associated RNase 1 (Ear1). Ear1 was also detected in airway epithelium and was reduced in lungs of mice exposed to neonatal hyperoxia. Electroporation-mediated gene delivery of Ear1 to the lung before infection successfully reduced viral replication and leukocyte recruitment during infection. It also diminished the enhanced morbidity and mortality attributed to neonatal hyperoxia. These findings demonstrate that novel epithelial expression of Ear1 functions to limit influenza A virus infection, and its loss contributes to oxygen-associated epithelial injury and fibrosis after infection. People born prematurely may have defects in epithelial innate immunity that increase their risk for respiratory viral infections.


Subject(s)
Eosinophil-Derived Neurotoxin/metabolism , Epithelium/metabolism , Influenza A virus/physiology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Oxygen/pharmacology , Ribonucleases/metabolism , Aging/pathology , Air , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Animals , Animals, Newborn , Electroporation , Epithelium/drug effects , Epithelium/pathology , Epithelium/virology , Female , Gene Transfer Techniques , Hyperoxia/complications , Hyperoxia/pathology , Hyperoxia/virology , Influenza A virus/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/prevention & control
11.
Bioelectrochemistry ; 151: 108396, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36805203

ABSTRACT

We used voltage-clamp electroporation to obtain single-channel recordings of lipid pores and analyzed the idealized dwell-time sequences using Maximum-Likelihood Fitting. We observed traces with multiple current levels and determined whether they were a result of the presence of multiple pores or a single pore with multiple conductance states. We found that, within the same recording, the bilayer can have a single pore with multiple conductance states or multiple independent pores. Using high sampling rates (100 kHz) we were able to observe pores with 40 µs lifetimes, and in experiments using high-voltage pulses we observed the existence of long-lived fluctuations minutes after the removal of the electric field. These results come closer to reconciling the nanosecond lifetime pores in molecular dynamics simulations and the long-lived permeability of cells after electroporation.


Subject(s)
Electroporation , Lipid Bilayers , Electroporation/methods , Electroporation Therapies , Molecular Dynamics Simulation
12.
Sci Rep ; 13(1): 6792, 2023 04 26.
Article in English | MEDLINE | ID: mdl-37100889

ABSTRACT

Acute Lung Injury/Acute Respiratory Distress Syndrome (ALI/ARDS) is characterized by diffuse alveolar damage and significant edema accumulation, which is associated with impaired alveolar fluid clearance (AFC) and alveolar-capillary barrier disruption, leading to acute respiratory failure. Our previous data showed that electroporation-mediated gene delivery of the Na+, K+-ATPase ß1 subunit not only increased AFC, but also restored alveolar barrier function through upregulation of tight junction proteins, leading to treatment of LPS-induced ALI in mice. More importantly, our recent publication showed that gene delivery of MRCKα, the downstream effector of ß1 subunit-mediated signaling towards upregulation of adhesive junctions and epithelial and endothelial barrier integrity, also provided therapeutic potential for ARDS treatment in vivo but without necessarily accelerating AFC, indicating that for ARDS treatment, improving alveolar capillary barrier function may be of more benefit than improving fluid clearance. In the present study, we investigated the therapeutical potential of ß2 and ß3 subunits, the other two ß isoforms of Na+, K+-ATPase, for LPS-induced ALI. We found that gene transfer of either the ß1, ß2, or ß3 subunits significantly increased AFC compared to the basal level in naïve animals and each gave similar increased AFC to each other. However, unlike that of the ß1 subunit, gene transfer of the ß2 or ß3 subunit into pre-injured animal lungs failed to show the beneficial effects of attenuated histological damage, neutrophil infiltration, overall lung edema, or increased lung permeability, indicating that ß2 or ß3 gene delivery could not treat LPS induced lung injury. Further, while ß1 gene transfer increased levels of key tight junction proteins in the lungs of injured mice, that of either the ß2 or ß3 subunit had no effect on levels of tight junction proteins. Taken together, this strongly suggests that restoration of alveolar-capillary barrier function alone may be of equal or even more benefit than improving AFC for ALI/ARDS treatment.


Subject(s)
Acute Lung Injury , Respiratory Distress Syndrome , Mice , Animals , Up-Regulation , Lipopolysaccharides/pharmacology , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Lung/pathology , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/therapy , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/genetics , Respiratory Distress Syndrome/therapy , Genetic Therapy , Tight Junction Proteins/metabolism , Pulmonary Alveoli/metabolism
13.
Bioelectrochemistry ; 147: 108162, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35691267

ABSTRACT

Current-Clamp electroporation refers to the application of a constant current across a membrane which results in voltage fluctuations due to the creation of electropores. This method allows for the measurement of electroporation across a long timescale (minutes) and facilitates the comparison between experimental and theoretical studies. Of particular interest is the claim in the literature that current-clamp electroporation results in the creation of a single pore. We simulated current-clamp electroporation using the Smoluchowski and Langevin equations and identified two possible mechanisms to explain the observed voltage fluctuations. The voltage fluctuations may be due to a single pore or a few pores growing and shrinking via a negative feedback mechanism or the opening and closing of pores in a larger population of pores. Our results suggest that current-clamp conditions do not necessarily result in the creation of a single pore. Additionally, we showed that the Langevin model is more accurate than the Smoluchowski model under conditions where there are only a few pores.


Subject(s)
Electroporation , Models, Theoretical , Computer Simulation , Electroporation/methods
14.
Biotechnol Bioeng ; 108(2): 446-53, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20830684

ABSTRACT

Cytoplasmic transport of large molecules such as plasmid DNA (pDNA) has been shown to increase when cells are subjected to mild levels of cyclic stretch for brief periods. In the case of pDNA, this is in part due to the increased active transport of pDNA along stabilized, acetylated microtubules in the cytoplasm, whose levels are increased in response to stretch. It also has been shown that disruption of the dense actin network leads to increased pDNA and macromolecule diffusion as well. We hypothesize that stretch not only increases active transport of pDNA but also, similar to actin disrupting drugs, decreases cytoplasmic stiffness leading to a less restive pathway for macromolecules to diffuse. To test this we used particle tracking microrheology to measure cytoplasmic mechanics. We conclude that while cyclic stretch transiently decreases cytoplasmic stiffness and increases diffusivity, stretch-independent modulation of the levels of acetylated, stable microtubules has no effect on cytoplasmic stiffness. Furthermore, stretching cells that have maximally acetylated microtubules increases cytoplasmic trafficking of pDNA, without increasing levels of acetylated microtubules. These findings suggest that stretch-enhanced gene transfer may occur by two independent mechanisms: increased levels of acetylated microtubules for directed active transport, and reduced cytoplasmic stiffness for increased diffusion.


Subject(s)
Cytoskeleton/metabolism , Epithelial Cells/physiology , Pulmonary Alveoli/cytology , Stress, Physiological , Cytoplasm/chemistry , Diffusion , Microtubules/metabolism , Plasmids/metabolism , Rheology/methods
15.
Sci Rep ; 11(1): 20862, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34675326

ABSTRACT

Acute Lung Injury/Acute Respiratory Distress Syndrome (ALI/ARDS) is characterized by alveolar edema accumulation with reduced alveolar fluid clearance (AFC), alveolar-capillary barrier disruption, and substantial inflammation, all leading to acute respiratory failure. Enhancing AFC has long been considered one of the primary therapeutic goals in gene therapy treatments for ARDS. We previously showed that electroporation-mediated gene delivery of the Na+, K+-ATPase ß1 subunit not only increased AFC, but also restored alveolar barrier function through upregulation of tight junction proteins, leading to treatment of LPS-induced ALI in mice. We identified MRCKα as an interaction partner of ß1 which mediates this upregulation in cultured alveolar epithelial cells. In this study, we investigate whether electroporation-mediated gene transfer of MRCKα to the lungs can attenuate LPS-induced acute lung injury in vivo. Compared to mice that received a non-expressing plasmid, those receiving the MRCKα plasmid showed attenuated LPS-increased pulmonary edema and lung leakage, restored tight junction protein expression, and improved overall outcomes. Interestingly, gene transfer of MRCKα did not alter AFC rates. Studies using both cultured microvascular endothelial cells and mice suggest that ß1 and MRCKα upregulate junctional complexes in both alveolar epithelial and capillary endothelial cells, and that one or both barriers may be positively affected by our approach. Our data support a model of treatment for ALI/ARDS in which improvement of alveolar-capillary barrier function alone may be of more benefit than improvement of alveolar fluid clearance.


Subject(s)
Acute Lung Injury/genetics , Gene Transfer Techniques , Myotonin-Protein Kinase/genetics , Up-Regulation , Acute Lung Injury/etiology , Acute Lung Injury/pathology , Acute Lung Injury/therapy , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Animals , Cell Line , Endothelial Cells , Genetic Therapy , Humans , Lipopolysaccharides/adverse effects , Male , Mice , Protein Serine-Threonine Kinases/genetics , Pulmonary Alveoli/blood supply , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology
16.
Front Physiol ; 12: 786255, 2021.
Article in English | MEDLINE | ID: mdl-35111077

ABSTRACT

Acute respiratory distress syndrome (ARDS) is a devastating clinical syndrome that leads to acute respiratory failure and accounts for over 70,000 deaths per year in the United States alone, even prior to the COVID-19 pandemic. While its molecular details have been teased apart and its pathophysiology largely established over the past 30 years, relatively few pharmacological advances in treatment have been made based on this knowledge. Indeed, mortality remains very close to what it was 30 years ago. As an alternative to traditional pharmacological approaches, gene therapy offers a highly controlled and targeted strategy to treat the disease at the molecular level. Although there is no single gene or combination of genes responsible for ARDS, there are a number of genes that can be targeted for upregulation or downregulation that could alleviate many of the symptoms and address the underlying mechanisms of this syndrome. This review will focus on the pathophysiology of ARDS and how gene therapy has been used for prevention and treatment. Strategies for gene delivery to the lung, such as barriers encountered during gene transfer, specific classes of genes that have been targeted, and the outcomes of these approaches on ARDS pathogenesis and resolution will be discussed.

17.
Exp Biol Med (Maywood) ; 246(3): 260-274, 2021 02.
Article in English | MEDLINE | ID: mdl-33183069

ABSTRACT

Delivery of genetic material to tissues in vivo is an important technique used in research settings and is the foundation upon which clinical gene therapy is built. The lung is a prime target for gene delivery due to a host of genetic, acquired, and infectious diseases that manifest themselves there, resulting in many pathologies. However, the in vivo delivery of genetic material to the lung remains a practical problem clinically and is considered the major obstacle needed to be overcome for gene therapy. Currently there are four main strategies for in vivo gene delivery to the lung: viral vectors, liposomes, nanoparticles, and electroporation. Viral delivery uses several different genetically modified viruses that enter the cell and express desired genes that have been inserted to the viral genome. Liposomes use combinations of charged and neutral lipids that can encapsulate genetic cargo and enter cells through endogenous mechanisms, thereby delivering their cargoes. Nanoparticles are defined by their size (typically less than 100 nm) and are made up of many different classes of building blocks, including biological and synthetic polymers, cell penetrant and other peptides, and dendrimers, that also enter cells through endogenous mechanisms. Electroporation uses mild to moderate electrical pulses to create pores in the cell membrane through which delivered genetic material can enter a cell. An emerging fifth category, exosomes and extracellular vesicles, may have advantages of both viral and non-viral approaches. These extracellular vesicles bud from cellular membranes containing receptors and ligands that may aid cell targeting and which can be loaded with genetic material for efficient transfer. Each of these vectors can be used for different gene delivery applications based on mechanisms of action, side-effects, and other factors, and their use in the lung and possible clinical considerations is the primary focus of this review.


Subject(s)
Gene Transfer Techniques , Genetic Therapy/methods , Lung Diseases/genetics , Lung Diseases/therapy , Electroporation/methods , Exosomes/genetics , Extracellular Vesicles/genetics , Humans , Liposomes/therapeutic use , Lung , Nanoparticles/therapeutic use , Viruses/genetics
18.
Methods Mol Biol ; 2208: 49-67, 2021.
Article in English | MEDLINE | ID: mdl-32856255

ABSTRACT

The potential of RNAi therapies has been largely impeded by the inherent challenges in the functional delivery of siRNA to cells. Herein, we describe protocols for the synthesis and characterization of novel peptide-siRNA nanoparticles prepared from disulfide-constrained amphipathic peptides complexed with siRNA as promising siRNA delivery vectors. We also describe protocols for the application of these nanoparticles to the in vitro and in vivo delivery of siRNA to lung cells for the functional knockdown of lung proteins.


Subject(s)
Disulfides/chemistry , Drug Delivery Systems/methods , Lung/drug effects , Nanoparticles/chemistry , Oligonucleotides/chemistry , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/chemistry , RNA, Small Interfering/chemistry , A549 Cells , Cell Line, Tumor , Gene Transfer Techniques , Humans , RNA Interference/physiology
19.
JCI Insight ; 6(4)2021 02 22.
Article in English | MEDLINE | ID: mdl-33507884

ABSTRACT

An intact lung epithelial barrier is essential for lung homeostasis. The Na+, K+-ATPase (NKA), primarily serving as an ion transporter, also regulates epithelial barrier function via modulation of tight junctions. However, the underlying mechanism is not well understood. Here, we show that overexpression of the NKA ß1 subunit upregulates the expression of tight junction proteins, leading to increased alveolar epithelial barrier function by an ion transport-independent mechanism. Using IP and mass spectrometry, we identified a number of unknown protein interactions of the ß1 subunit, including a top candidate, myotonic dystrophy kinase-related cdc42-binding kinase α (MRCKα), which is a protein kinase known to regulate peripheral actin formation. Using a doxycycline-inducible gene expression system, we demonstrated that MRCKα and its downstream activation of myosin light chain is required for the regulation of alveolar barrier function by the NKA ß1 subunit. Importantly, MRCKα is expressed in both human airways and alveoli and has reduced expression in patients with acute respiratory distress syndrome (ARDS), a lung illness that can be caused by multiple direct and indirect insults, including the infection of influenza virus and SARS-CoV-2. Our results have elucidated a potentially novel mechanism by which NKA regulates epithelial tight junctions and have identified potential drug targets for treating ARDS and other pulmonary diseases that are caused by barrier dysfunction.


Subject(s)
Myotonin-Protein Kinase/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Tight Junctions/metabolism , Alveolar Epithelial Cells/cytology , Alveolar Epithelial Cells/metabolism , Animals , HEK293 Cells , Humans , Myotonin-Protein Kinase/genetics , Primary Cell Culture , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Respiratory Distress Syndrome/pathology , Respiratory Distress Syndrome/virology , SARS-CoV-2/pathogenicity , Sodium-Potassium-Exchanging ATPase/genetics
20.
Sci Rep ; 11(1): 1453, 2021 01 14.
Article in English | MEDLINE | ID: mdl-33446928

ABSTRACT

Exogenous electric fields are currently used in human therapy in a number of contexts. Interestingly, electric fields have also been shown to alter migration and function of immune cells, suggesting the potential for electric field-based immune therapy. Little is known as to the effect of electric field treatment (EFT) on the lung. To determine if EFT associates with changes in lung immune cell infiltration, we used a mouse model with varying methods of EFT application and measured cells and soluble mediators using flow cytometry and cytokine/chemokine multiplex. EFT was associated with a transient increase in lung neutrophils and decrease in eosinophils in naïve mice within 2 h of treatment, accompanied by an increase in IL-6 levels. In order to test whether EFT could alter eosinophil/neutrophil recruitment in a relevant disease model, a mouse model of allergic airway inflammation was used. Four EFT doses in allergen-sensitized mice resulted in increased neutrophil and reduced eosinophil infiltrates following allergen challenge, suggesting a durable change in inflammation by EFT. Mice with allergic inflammation were analyzed by flexiVent for measures of lung function. EFT-treated mice had increased inspiratory capacity and other measures of lung function were not diminished. These data suggest EFT may be used to manipulate immune cell infiltration in the lung without affecting lung function.


Subject(s)
Asthma/immunology , Electricity , Eosinophils/immunology , Lung/immunology , Neutrophil Infiltration , Neutrophils/immunology , Animals , Asthma/pathology , Eosinophils/pathology , Lung/pathology , Mice , Neutrophils/pathology
SELECTION OF CITATIONS
SEARCH DETAIL