Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 122
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 181(5): 955-960, 2020 05 28.
Article in English | MEDLINE | ID: mdl-32470403

ABSTRACT

The first clinical studies utilizing RNA-guided endonucleases (RGENs) to therapeutically edit RNA and DNA in cancer patients were recently published. These groundbreaking technological advances promise to revolutionize genetic therapy and, as I discuss, represent the culmination of decades of innovative work to engineer RGENs for such editing applications.


Subject(s)
Gene Editing/methods , Gene Editing/trends , RNA Editing/genetics , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , DNA/genetics , Endonucleases/metabolism , Mutation , RNA/genetics , RNA Editing/physiology , RNA, Catalytic/genetics , RNA, Guide, Kinetoplastida/genetics
2.
RNA ; 29(4): 455-462, 2023 04.
Article in English | MEDLINE | ID: mdl-36697262

ABSTRACT

In this short Perspective, we discuss the history of, and recent progress toward, the development of aptamers that can serve as rapid onset anticoagulants during cardiopulmonary bypass (CPB), extracorporeal membrane oxygenation (ECMO), and catheter-based diagnostic and interventional procedures, several million of which are performed each year worldwide. Aptamer anticoagulants provide potent and antidote-controllable anticoagulation and have low immunogenicity. New methods of aptamer isolation and engineering have not only improved the quality of aptamers, but also accelerated their development. Unfortunately, no aptamer identified to date can produce an anticoagulant effect as potent as that produced by unfractionated heparin (UFH), the standard anticoagulant for CPB. We have suggested several possible strategies to amplify the anticoagulant potency of existing aptamer anticoagulants.


Subject(s)
Aptamers, Nucleotide , Heparin , Heparin/pharmacology , Aptamers, Nucleotide/pharmacology , Aptamers, Nucleotide/therapeutic use , Blood Coagulation , Anticoagulants/pharmacology , Anticoagulants/therapeutic use , Antidotes/pharmacology
3.
Mol Ther ; 30(2): 845-854, 2022 02 02.
Article in English | MEDLINE | ID: mdl-34628051

ABSTRACT

Nucleic acid (NA)-containing damage- and pathogen-associated molecular patterns (DAMPs and PAMPs, respectively) are implicated in numerous pathological conditions from infectious diseases to autoimmune disorders. Nucleic acid-binding polymers, including polyamidoamine (PAMAM) dendrimers, have demonstrated anti-inflammatory properties when administered to neutralize DAMPs/PAMPs. The PAMAM G3 variant has been shown to have beneficial effects in a cutaneous lupus erythematosus (CLE) murine model and improve survival of mice challenged with influenza. Unfortunately, the narrow therapeutic window of cationic PAMAM dendrimers makes their clinical development challenging. An alternative nucleic acid-binding polymer that has been evaluated in humans is a linear ß-cyclodextrin-containing polymer (CDP). CDP's characteristics prompted us to evaluate its anti-inflammatory potential in CLE autoimmune and influenza infectious disease mouse models. We report that CDP effectively inhibits NA-containing DAMP-mediated activation of Toll-like receptors (TLRs) in cell culture, improves healing in lupus mice, and does not immunocompromise treated animals upon influenza infection but improves survival even when administered 3 days after infection. Finally, as anticipated, we observe limited toxicity in animals treated with CDP compared with PAMAM G3. Thus, CDP is a new anti-inflammatory agent that may be readily translated to the clinic to combat diseases associated with pathological NA-containing DAMPs/PAMPs.


Subject(s)
Influenza, Human , Lupus Erythematosus, Cutaneous , Nucleic Acids , beta-Cyclodextrins , Animals , Humans , Lupus Erythematosus, Cutaneous/drug therapy , Mice , Nucleic Acids/chemistry , Polymers , beta-Cyclodextrins/therapeutic use
4.
Anesthesiology ; 137(1): 67-78, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35412597

ABSTRACT

BACKGROUND: COVID-19 causes hypercoagulability, but the association between coagulopathy and hypoxemia in critically ill patients has not been thoroughly explored. This study hypothesized that severity of coagulopathy would be associated with acute respiratory distress syndrome severity, major thrombotic events, and mortality in patients requiring intensive care unit-level care. METHODS: Viscoelastic testing by rotational thromboelastometry and coagulation factor biomarker analyses were performed in this prospective observational cohort study of critically ill COVID-19 patients from April 2020 to October 2020. Statistical analyses were performed to identify significant coagulopathic biomarkers such as fibrinolysis-inhibiting plasminogen activator inhibitor 1 and their associations with clinical outcomes such as mortality, extracorporeal membrane oxygenation requirement, occurrence of major thrombotic events, and severity of hypoxemia (arterial partial pressure of oxygen/fraction of inspired oxygen categorized into mild, moderate, and severe per the Berlin criteria). RESULTS: In total, 53 of 55 (96%) of the cohort required mechanical ventilation and 9 of 55 (16%) required extracorporeal membrane oxygenation. Extracorporeal membrane oxygenation-naïve patients demonstrated lysis indices at 30 min indicative of fibrinolytic suppression on rotational thromboelastometry. Survivors demonstrated fewer procoagulate acute phase reactants, such as microparticle-bound tissue factor levels (odds ratio, 0.14 [0.02, 0.99]; P = 0.049). Those who did not experience significant bleeding events had smaller changes in ADAMTS13 levels compared to those who did (odds ratio, 0.05 [0, 0.7]; P = 0.026). Elevations in plasminogen activator inhibitor 1 (odds ratio, 1.95 [1.21, 3.14]; P = 0.006), d-dimer (odds ratio, 3.52 [0.99, 12.48]; P = 0.05), and factor VIII (no clot, 1.15 ± 0.28 vs. clot, 1.42 ± 0.31; P = 0.003) were also demonstrated in extracorporeal membrane oxygenation-naïve patients who experienced major thrombotic events. Plasminogen activator inhibitor 1 levels were significantly elevated during periods of severe compared to mild and moderate acute respiratory distress syndrome (severe, 44.2 ± 14.9 ng/ml vs. mild, 31.8 ± 14.7 ng/ml and moderate, 33.1 ± 15.9 ng/ml; P = 0.029 and 0.039, respectively). CONCLUSIONS: Increased inflammatory and procoagulant markers such as plasminogen activator inhibitor 1, microparticle-bound tissue factor, and von Willebrand factor levels are associated with severe hypoxemia and major thrombotic events, implicating fibrinolytic suppression in the microcirculatory system and subsequent micro- and macrovascular thrombosis in severe COVID-19.


Subject(s)
Blood Coagulation Disorders , COVID-19 , Respiratory Distress Syndrome , Thrombophilia , Thrombosis , Blood Coagulation Disorders/complications , COVID-19/complications , Critical Illness , Fibrinolysis , Humans , Hypoxia/complications , Microcirculation , Oxygen , Plasminogen Activator Inhibitor 1 , Prospective Studies , Retrospective Studies , Thrombophilia/complications , Thromboplastin
5.
Proc Natl Acad Sci U S A ; 115(18): 4761-4766, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29666232

ABSTRACT

Therapies that can eliminate both local and metastatic prostate tumor lesions while sparing normal organ tissue are desperately needed. With the goal of developing an improved drug-targeting strategy, we turned to a new class of targeted anticancer therapeutics: aptamers conjugated to highly toxic chemotherapeutics. Cell selection for aptamers with prostate cancer specificity yielded the E3 aptamer, which internalizes into prostate cancer cells without targeting normal prostate cells. Chemical conjugation of E3 to the drugs monomethyl auristatin E (MMAE) and monomethyl auristatin F (MMAF) yields a potent cytotoxic agent that efficiently kills prostate cancer cells in vitro but does not affect normal prostate epithelial cells. Importantly, the E3 aptamer targets tumors in vivo and treatment with the MMAF-E3 conjugate significantly inhibits prostate cancer growth in mice, demonstrating the in vivo utility of aptamer-drug conjugates. Additionally, we report the use of antidotes to block E3 aptamer-drug conjugate cytotoxicity, providing a safety switch in the unexpected event of normal cell killing in vivo.


Subject(s)
Aminobenzoates/pharmacology , Antineoplastic Agents/pharmacology , Aptamers, Nucleotide/pharmacology , Oligopeptides/pharmacology , Prostatic Neoplasms/drug therapy , Aminobenzoates/chemistry , Animals , Antineoplastic Agents/chemistry , Aptamers, Nucleotide/chemistry , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacology , Humans , Male , Mice , Mice, Nude , Oligopeptides/chemistry , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Xenograft Model Antitumor Assays
6.
Annu Rev Pharmacol Toxicol ; 57: 61-79, 2017 01 06.
Article in English | MEDLINE | ID: mdl-28061688

ABSTRACT

Aptamers are single-stranded nucleic acid molecules that bind to and inhibit proteins and are commonly produced by systematic evolution of ligands by exponential enrichment (SELEX). Aptamers undergo extensive pharmacological revision, which alters affinity, specificity, and therapeutic half-life, tailoring each drug for a specific clinical need. The first therapeutic aptamer was described 25 years ago. Thus far, one aptamer has been approved for clinical use, and numerous others are in preclinical or clinical development. This review presents a short history of aptamers and SELEX, describes their pharmacological development and optimization, and reviews potential treatment of diseases including visual disorders, thrombosis, and cancer.


Subject(s)
Aptamers, Nucleotide/administration & dosage , Aptamers, Nucleotide/genetics , SELEX Aptamer Technique/methods , Animals , Aptamers, Nucleotide/metabolism , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Randomized Controlled Trials as Topic/methods , SELEX Aptamer Technique/trends , Vision Disorders/drug therapy , Vision Disorders/metabolism
7.
Mol Ther ; 27(7): 1228-1241, 2019 07 03.
Article in English | MEDLINE | ID: mdl-30987839

ABSTRACT

Endothelial surface and circulating glycoprotein von Willebrand factor (vWF) regulates platelet adhesion and is associated with thrombotic diseases, including ischemic stroke, myocardial infarction, and peripheral vascular disease. Thrombosis, as manifested in these diseases, is the leading cause of disability and death in the western world. Current parenteral antithrombotic and thrombolytic agents used to treat these conditions are limited by a short therapeutic window, irreversibility, and major risk of hemorrhage. To overcome these limitations, we developed a novel anti-vWF aptamer, called DTRI-031, that selectively binds and inhibits vWF-mediated platelet adhesion and arterial thrombosis while enabling rapid reversal of this antiplatelet activity by an antidote oligonucleotide (AO). Aptamer DTRI-031 exerts dose-dependent inhibition of platelet aggregation and thrombosis in whole blood and mice, respectively. Moreover, DTRI-031 can achieve potent vascular recanalization of platelet-rich thrombotic occlusions in murine and canine carotid arteries. Finally, DTRI-031 activity is rapidly (<5 min) and completely reversed by AO administration in a murine saphenous vein hemorrhage model, and murine toxicology studies indicate the aptamer is well tolerated. These findings suggest that targeting vWF with an antidote-controllable aptamer potentially represents an effective and safer treatment for thrombosis patients having platelet-rich arterial occlusions in the brain, heart, or periphery.


Subject(s)
Aptamers, Nucleotide/pharmacology , Arterial Occlusive Diseases/drug therapy , Drug Evaluation, Preclinical/methods , Fibrinolytic Agents/pharmacology , Thrombosis/drug therapy , Thrombosis/prevention & control , von Willebrand Factor/antagonists & inhibitors , Animals , Antidotes/pharmacology , Aptamers, Nucleotide/chemical synthesis , Aptamers, Nucleotide/metabolism , Blood Platelets/drug effects , Blood Platelets/metabolism , Carotid Artery Injuries/drug therapy , Dogs , Dose-Response Relationship, Drug , Female , Healthy Volunteers , Humans , Male , Mice , Mice, Inbred C57BL , Oligonucleotides/pharmacology , Platelet Adhesiveness/drug effects , Platelet Aggregation/drug effects , von Willebrand Factor/metabolism
8.
Mol Ther ; 26(4): 1020-1031, 2018 04 04.
Article in English | MEDLINE | ID: mdl-29550075

ABSTRACT

Nucleic acid binding polymers (NABPs) have been extensively used as vehicles for DNA and RNA delivery. More recently, we discovered that a subset of these NABPs can also serve as anti-inflammatory agents by capturing pro-inflammatory extracellular nucleic acids and associated protein complexes that promote activation of toll-like receptors (TLRs) in diseases such as lupus erythematosus. Nucleic-acid-mediated TLR signaling also facilitates tumor progression and metastasis in several cancers, including pancreatic cancer (PC). In addition, extracellular DNA and RNA circulate on or within lipid microvesicles, such as microparticles or exosomes, which also promote metastasis by inducing pro-tumorigenic signaling in cancer cells and pre-conditioning secondary sites for metastatic establishment. Here, we explore the use of an NABP, the 3rd generation polyamidoamine dendrimer (PAMAM-G3), as an anti-metastatic agent. We show that PAMAM-G3 not only inhibits nucleic-acid-mediated activation of TLRs and invasion of PC tumor cells in vitro, but can also directly bind extracellular microvesicles to neutralize their pro-invasive effects as well. Moreover, we demonstrate that PAMAM-G3 dramatically reduces liver metastases in a syngeneic murine model of PC. Our findings identify a promising therapeutic application of NABPs for combating metastatic disease in PC and potentially other malignancies.


Subject(s)
Alarmins/metabolism , Cell-Derived Microparticles/metabolism , Nucleic Acids/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Polymers , Animals , Cell Line, Tumor , Dendrimers/chemistry , Dendrimers/metabolism , Disease Models, Animal , Exosomes/metabolism , Humans , Mice , Neoplasm Invasiveness , Neoplasm Staging , Nucleic Acids/chemistry , Pancreatic Neoplasms/therapy , Polymers/chemistry , Polymers/metabolism , Protein Binding , Toll-Like Receptor 9/metabolism
9.
Proc Natl Acad Sci U S A ; 113(35): 9728-33, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27528673

ABSTRACT

Nucleic acid-containing debris released from dead and dying cells can be recognized as damage-associated molecular patterns (DAMPs) or pattern-associated molecular patterns (PAMPs) by the innate immune system. Inappropriate activation of the innate immune response can engender pathological inflammation and autoimmune disease. To combat such diseases, major efforts have been made to therapeutically target the pattern recognition receptors (PRRs) such as the Toll-like receptors (TLRs) that recognize such DAMPs and PAMPs, or the downstream effector molecules they engender, to limit inflammation. Unfortunately, such strategies can limit the ability of the immune system to combat infection. Previously, we demonstrated that nucleic acid-binding polymers can act as molecular scavengers and limit the ability of artificial nucleic acid ligands to activate PRRs. Herein, we demonstrate that nucleic acid scavengers (NASs) can limit pathological inflammation and nucleic acid-associated autoimmunity in lupus-prone mice. Moreover, we observe that such NASs do not limit an animal's ability to combat viral infection, but rather their administration improves survival when animals are challenged with lethal doses of influenza. These results indicate that molecules that scavenge extracellular nucleic acid debris represent potentially safer agents to control pathological inflammation associated with a wide range of autoimmune and infectious diseases.


Subject(s)
Antibodies, Antinuclear/metabolism , Dendrimers/pharmacology , Immunologic Factors/pharmacology , Lupus Erythematosus, Cutaneous/drug therapy , Nucleic Acids/isolation & purification , Skin/drug effects , Animals , Autoimmunity/drug effects , DNA Cleavage , Humans , Lupus Erythematosus, Cutaneous/immunology , Lupus Erythematosus, Cutaneous/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nucleic Acids/chemistry , Protein Binding , RNA Cleavage , Skin/immunology , Skin/pathology
10.
Curr Opin Hematol ; 25(5): 382-388, 2018 09.
Article in English | MEDLINE | ID: mdl-30015643

ABSTRACT

PURPOSE OF REVIEW: Since the selection of the first thrombin-binding aptamer in 1992, the use of nucleic acid aptamers to target specific coagulation factors has emerged as a valuable approach for generating novel anticoagulant and procoagulant therapeutics. Herein, we highlight the most recent discoveries involving application of aptamers for those purposes. RECENT FINDINGS: Learning from the successes and pitfalls of the FIXa-targeting aptamer pegnivacogin in preclinical and clinical studies, the latest efforts to develop antidote-controllable anticoagulation strategies for cardiopulmonary bypass that avoid unfractionated heparin involve potentiation of the exosite-binding factor X (FX)a aptamer 11F7t by combination with either a small molecule FXa catalytic site inhibitor or a thrombin aptamer. Recent work has also focused on identifying aptamer inhibitors of contact pathway factors such as FXIa and kallikrein, which may prove to be well tolerated and effective antithrombotic agents in certain clinical settings. Finally, new approaches to develop procoagulant aptamers to control bleeding associated with hemophilia and other coagulopathies involve targeting activated protein C and tissue plasminogen activator. SUMMARY: Overall, these recent findings exemplify the versatility of aptamers to modulate a variety of procoagulant and anticoagulant factors, along with their capacity to be used complementarily with other aptamers or drugs for wide-ranging applications.


Subject(s)
Aptamers, Nucleotide/therapeutic use , Factor IXa , Factor Xa Inhibitors/therapeutic use , Hemostasis , Animals , Antidotes/pharmacokinetics , Antidotes/therapeutic use , Aptamers, Nucleotide/adverse effects , Aptamers, Nucleotide/pharmacokinetics , Cardiopulmonary Bypass , Catalytic Domain , Factor IXa/antagonists & inhibitors , Factor IXa/metabolism , Factor Xa Inhibitors/adverse effects , Factor Xa Inhibitors/pharmacokinetics , Hemophilia A/blood , Hemophilia A/drug therapy , Humans , Kallikreins/metabolism
11.
Nat Chem Biol ; 12(9): 709-16, 2016 09.
Article in English | MEDLINE | ID: mdl-27398998

ABSTRACT

G-protein-coupled receptor (GPCR) ligands function by stabilizing multiple, functionally distinct receptor conformations. This property underlies the ability of 'biased agonists' to activate specific subsets of a given receptor's signaling profile. However, stabilizing distinct active GPCR conformations to enable structural characterization of mechanisms underlying GPCR activation remains difficult. These challenges have accentuated the need for receptor tools that allosterically stabilize and regulate receptor function through unique, previously unappreciated mechanisms. Here, using a highly diverse RNA library combined with advanced selection strategies involving state-of-the-art next-generation sequencing and bioinformatics analyses, we identify RNA aptamers that bind a prototypical GPCR, the ß2-adrenoceptor (ß2AR). Using biochemical, pharmacological, and biophysical approaches, we demonstrate that these aptamers bind with nanomolar affinity at defined surfaces of the receptor, allosterically stabilizing active, inactive, and ligand-specific receptor conformations. The discovery of RNA aptamers as allosteric GPCR modulators significantly expands the diversity of ligands available to study the structural and functional regulation of GPCRs.


Subject(s)
Aptamers, Nucleotide/metabolism , Receptors, Adrenergic, beta-2/metabolism , Allosteric Regulation/drug effects , Aptamers, Nucleotide/chemistry , Benzoxazines/chemistry , Benzoxazines/pharmacology , Humans , Models, Molecular , Protein Conformation , Receptors, Adrenergic, beta-2/chemistry
12.
J Surg Res ; 231: 270-277, 2018 11.
Article in English | MEDLINE | ID: mdl-30278940

ABSTRACT

BACKGROUND: Surgical insult and trauma have been shown to cause dysregulation of the immune and inflammatory responses. Interaction of damage-associated molecular patterns (DAMPs) with toll-like receptors (TLRs) initiates innate immune response and systemic inflammatory responses. Given that surgical patients produce high levels of circulating damage-associated molecular patterns, we hypothesized that plasma-activated TLR activity would be correlated to injury status and could be used to predict pathological conditions involving tissue injury. METHODS: An observational study was performed using samples from a single-institution prospective tissue and data repository from a Level-1 trauma center. In vitro TLR 2, 3, 4, and 9 activation was determined in a TLR reporter assay after isolation of plasma from peripheral blood. We determined correlations between plasma-activated TLR activity and clinical course measures of severity. RESULTS: Eighteen patients were enrolled (median Injury Severity Score 15 [interquartile range 10, 23.5]). Trauma resulted in significant elevation in circulation high mobility group box 1 as well as increase of plasma-activated TLR activation (2.8-5.4-fold) compared to healthy controls. There was no correlation between circulating high mobility group box 1 and trauma morbidity; however, the plasma-activated TLR activity was correlated with acute physiology and chronic health evaluation II scores (R square = 0.24-0.38, P < 0.05). Patients who received blood products demonstrated significant increases in the levels of plasma-activated TLRs 2, 3, 4, and 9 and had a trend toward developing systemic inflammatory response syndrome. CONCLUSIONS: Further studies examining TLR modulation and signaling in surgical patients may assist in predictive risk modeling and reduction in morbidity and mortality.


Subject(s)
Alarmins/metabolism , Toll-Like Receptors/blood , Wounds and Injuries/blood , Adult , Biomarkers/blood , Case-Control Studies , Cell Line, Tumor , Female , HMGB1 Protein/blood , Humans , Male , Middle Aged
13.
Mol Ther ; 25(6): 1295-1305, 2017 06 07.
Article in English | MEDLINE | ID: mdl-28372998

ABSTRACT

Activation of the RNA-sensing pattern recognition receptor (PRR) in cancer cells leads to cell death and cytokine expression. This cancer cell death releases tumor antigens and damage-associated molecular patterns (DAMPs) that induce anti-tumor immunity. However, these cytokines and DAMPs also cause adverse inflammatory and thrombotic complications that can limit the overall therapeutic benefits of PRR-targeting anti-cancer therapies. To overcome this problem, we generated and evaluated two novel and distinct ssRNA molecules (immunogenic cell-killing RNA [ICR]2 and ICR4). ICR2 and ICR4 differentially stimulated cell death and PRR signaling pathways and induced different patterns of cytokine expression in cancer and innate immune cells. Interestingly, DAMPs released from ICR2- and ICR4-treated cancer cells had distinct patterns of stimulation of innate immune receptors and coagulation. Finally, ICR2 and ICR4 inhibited in vivo tumor growth as effectively as poly(I:C). ICR2 and ICR4 are potential therapeutic agents that differentially induce cell death, immune stimulation, and coagulation when introduced into tumors.


Subject(s)
Cell Differentiation/genetics , Gene Expression Regulation, Neoplastic , Interferons/genetics , Neoplasms/genetics , Neoplasms/immunology , RNA/genetics , Receptors, Pattern Recognition/genetics , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Calbindin 2/metabolism , Cell Death/genetics , Cell Death/immunology , Cell Line, Tumor , Cytokines/genetics , Disease Models, Animal , HMGB1 Protein/metabolism , Heterografts , Humans , Immunity, Innate/genetics , Inflammation Mediators , Mice , Neoplasms/metabolism , Protein Transport , RNA/chemistry , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/genetics , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Receptors, Pattern Recognition/metabolism , Signal Transduction
14.
Biochem Biophys Res Commun ; 478(3): 1484-90, 2016 09 23.
Article in English | MEDLINE | ID: mdl-27586271

ABSTRACT

Transfection with in vitro transcribed mRNAs is a safe and effective tool to convert somatic cells to any cell type of interest. One caveat of mRNA transfection is that mRNAs are recognized by multiple RNA-sensing toll like receptors (TLRs). These TLRs can both promote and inhibit cellular reprogramming. We demonstrated that mRNA transfection stimulated TLR3 and TLR7 and induced cytotoxicity and IFN-ß expression in human and mouse fibroblasts. Furthermore, mRNA transfection induced paracrine inhibition of repeated mRNA transfection through type I IFNs. Modified mRNAs (mmRNAs) containing pseudouridine and 5-methycytosine reduced TLR stimulation, cytotoxicity and IFN-ß expression in fibroblasts. Repeated liposomal transfection with MyoD mmRNAs significantly enhanced myogenic conversion of human and mouse fibroblasts compared with repeated transfection with MyoD mRNAs. Interestingly, electroporation of mRNAs and mmRNAs completely abrogated cytotoxicity and IFN-ß expression and also abolished myogenic conversion of fibroblasts. At a low concentration, TLR7/8 agonist R848 enhanced MyoD mmRNA-driven conversion of human fibroblasts into skeletal muscle cells, whereas high concentrations of R848 inhibited myogenic conversion of fibroblasts. Our study suggests that deliberate control of TLR signaling is a key factor in the success of mRNA-driven cellular reprogramming.


Subject(s)
Fibroblasts/metabolism , Muscle Development , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 7/metabolism , Animals , Cell Death , Electroporation , Humans , Infant, Newborn , Interferon-beta/metabolism , Mice , Muscle Development/genetics , MyoD Protein/genetics , MyoD Protein/metabolism , Paracrine Communication , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Transcription, Genetic , Transfection
15.
Biomacromolecules ; 17(11): 3706-3713, 2016 11 14.
Article in English | MEDLINE | ID: mdl-27741396

ABSTRACT

Dying cells release nucleic acids (NA) and NA-containing complexes that activate inflammatory pathways of immune cells. Sustained activation of these pathways contributes to chronic inflammation frequently encountered in autoimmune and inflammatory diseases. In this study, grafting of cationic polymers onto a nanofibrous mesh enabled local scavenging of negatively charged pro-inflammatory molecules in the extracellular space. Nucleic acid scavenging nanofibers (NASFs) formed from poly(styrene-alt-maleic anhydride) conjugated with 1.8 kDa bPEI resulted in nanofibers of diameters 486 ± 9 nm. NASFs inhibited the NF-κB response stimulated by the negatively charged agonists, CpG and poly(I:C), in Ramos-blue cells but not Pam3CSK4, a nonanionic agonist. Moreover, NASFs significantly impeded NF-κB activation in cells stimulated with damage-associated molecular pattern molecules (DAMPs) released from doxorubicin killed cancer cells. In vivo application of NASFs to open wounds demonstrated nucleic acid scavenging in wounds of diabetic mice infected with Pseudomonas aeruginosa, suggesting the in vivo efficacy of NASFs. This simple technique of generating NASF results in effective localized anti-inflammation in vitro and local nucleic acid scavenging in vivo.


Subject(s)
Inflammation/drug therapy , Maleates/chemistry , Nanofibers/chemistry , Polystyrenes/chemistry , Wound Healing/drug effects , Animals , Doxorubicin/administration & dosage , Doxorubicin/chemistry , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Humans , Inflammation/microbiology , Inflammation/pathology , Maleates/administration & dosage , Mice , Mice, Inbred NOD , Nanofibers/administration & dosage , Nucleic Acids/chemistry , Polyamines/administration & dosage , Polyamines/chemistry , Polyelectrolytes , Polystyrenes/administration & dosage , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/pathogenicity
16.
Arterioscler Thromb Vasc Biol ; 35(10): 2083-91, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26315404

ABSTRACT

As a novel class of therapeutics, aptamers, or nucleic acid ligands, have garnered clinical interest because of the ease of isolating a highly specific aptamer against a wide range of targets, their chemical flexibility and synthesis, and their inherent ability to have their function reversed. The following review details the development and molecular mechanisms of aptamers targeting specific proteases in the coagulation cascade. The ability of these anticoagulant aptamers to bind to and inhibit exosite function rather than binding within the active site highlights the importance of exosites in blocking protein function. As both exosite inhibitors and reversible agents, the use of aptamers is a promising strategy for future therapeutics.


Subject(s)
Aptamers, Nucleotide/therapeutic use , Blood Coagulation/drug effects , Molecular Targeted Therapy/methods , Aptamers, Nucleotide/pharmacology , Blood Coagulation/genetics , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Female , Humans , Male , Prothrombin/drug effects , Prothrombin/metabolism , Sensitivity and Specificity , Serine Endopeptidases/drug effects , Serine Endopeptidases/metabolism , Thrombin/drug effects , Thrombin/metabolism
17.
Proc Natl Acad Sci U S A ; 109(32): 12938-43, 2012 Aug 07.
Article in English | MEDLINE | ID: mdl-22837404

ABSTRACT

Development of effective, yet safe, antithrombotic agents has been challenging because such agents increase the propensity of patients to bleed. Recently, naturally occurring polyphosphates such as extracellular DNA, RNA, and inorganic polyphosphates have been shown to activate blood coagulation. In this report, we evaluate the anticoagulant and antithrombotic activity of nucleic acid-binding polymers in vitro and in vivo. Such polymers bind to DNA, RNA, and inorganic polyphosphate molecules with high affinity and inhibit RNA- and polyphosphate-induced clotting and the activation of the intrinsic pathway of coagulation in vitro. Moreover, [NH(2)(CH(2))(2)NH(2)](G = 3);dendri PAMAM(NH(2))(32) (PAMAM G-3) prevents thrombosis following carotid artery injury and pulmonary thromboembolism in mice without significantly increasing blood loss from surgically challenged animals. These studies indicate that nucleic acid-binding polymers are able to scavenge effectively prothrombotic nucleic acids and other polyphosphates in vivo and represent a new and potentially safer class of antithrombotic agents.


Subject(s)
Blood Coagulation/drug effects , Fibrinolytic Agents/pharmacology , Nucleic Acids/metabolism , Polyphosphates/pharmacology , Thrombosis/drug therapy , Animals , Calorimetry , Carotid Artery Injuries/drug therapy , Mice , Pulmonary Embolism/drug therapy , Thrombelastography
18.
Proc Natl Acad Sci U S A ; 108(34): 14055-60, 2011 Aug 23.
Article in English | MEDLINE | ID: mdl-21844380

ABSTRACT

Dead and dying cells release nucleic acids. These extracellular RNAs and DNAs can be taken up by inflammatory cells and activate multiple nucleic acid-sensing toll-like receptors (TLR3, 7, 8, and 9). The inappropriate activation of these TLRs can engender a variety of inflammatory and autoimmune diseases. The redundancy of the TLR family encouraged us to seek materials that can neutralize the proinflammatory effects of any nucleic acid regardless of its sequence, structure or chemistry. Herein we demonstrate that certain nucleic acid-binding polymers can inhibit activation of all nucleic acid-sensing TLRs irrespective of whether they recognize ssRNA, dsRNA or hypomethylated DNA. Furthermore, systemic administration of such polymers can prevent fatal liver injury engendered by proinflammatory nucleic acids in an acute toxic shock model in mice. Therefore these polymers represent a novel class of anti-inflammatory agent that can act as molecular scavengers to neutralize the proinflammatory effects of various nucleic acids.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Nucleic Acids/metabolism , Polymers/pharmacology , Animals , Anti-Inflammatory Agents/therapeutic use , Cations , Cell Line , Endocytosis/drug effects , Extracellular Space/drug effects , Extracellular Space/metabolism , Humans , Immunity/drug effects , Inflammation/pathology , Intracellular Space/drug effects , Intracellular Space/immunology , Ligands , Liver Diseases/complications , Liver Diseases/drug therapy , Mice , Mice, Inbred C57BL , Nucleic Acids/toxicity , Oligodeoxyribonucleotides/pharmacology , Polymers/therapeutic use , Shock, Septic/complications , Shock, Septic/drug therapy , Toll-Like Receptors/immunology
19.
Nucleic Acid Ther ; 34(1): 12-17, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38285522

ABSTRACT

The ability to reverse the binding of aptamers to their target proteins has received considerable attention for developing controllable therapeutic agents. Recently, use of aptamers as reversible cell-sorting ligands has also sparked interest. Antibodies are currently utilized for isolating cells expressing a particular cell surface receptor. The inability to remove antibodies from isolated cells following sorting greatly limits their utility for many applications. Previously, we described how a particular aptamer-antidote oligonucleotide pair can isolate cells and clean them. Here, we demonstrate that this approach is generalizable; aptamers can simultaneously recognize more than one cell type during fluorescent activated cell sorting (FACS). Moreover, we describe a novel approach to reverse aptamer binding following cell sorting using a nuclease. This alternative strategy represents a cleaning approach that does not require the generation of antidote oligonucleotides for each aptamer and will greatly reduce the cost and expand the utility of Clean FACS.


Subject(s)
Antidotes , Aptamers, Nucleotide , Aptamers, Nucleotide/genetics , Aptamers, Nucleotide/pharmacology , Ligands , Staining and Labeling , Antibodies , SELEX Aptamer Technique
20.
Pain ; 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38452223

ABSTRACT

ABSTRACT: Secreted microRNAs (miRNAs) have been detected in various body fluids including the cerebrospinal fluid, yet their direct role in regulating synaptic transmission remains uncertain. We found that intrathecal injection of low dose of let-7b (1 µg) induced short-term (<24 hours) mechanical allodynia and heat hyperalgesia, a response that is compromised in Tlr7-/- or Trpa1-/- mice. Ex vivo and in vivo calcium imaging in GCaMP6-report mice revealed increased calcium signal in spinal cord afferent terminals and doral root ganglion/dorsal root ganglia neurons following spinal perfusion and intraplantar injection of let-7b. Patch-clamp recordings also demonstrated enhanced excitatory synaptic transmission (miniature excitatory postsynaptic currents [EPSCs]) in spinal nociceptive neurons following let-7b perfusion or optogenetic activation of axonal terminals. The elevation in spinal calcium signaling and EPSCs was dependent on the presence of toll-like receptor-7 (TLR7) and transient receptor potential ion channel subtype A1 (TRPA1). In addition, endogenous let-7b is enriched in spinal cord synaptosome, and peripheral inflammation increased let-7b in doral root ganglion/dorsal root ganglia neurons, spinal cord tissue, and the cerebrospinal fluid. Notably, let-7b antagomir inhibited inflammatory pain and inflammation-induced synaptic plasticity (EPSC increase), suggesting an endogenous role of let-7b in regulating pain and synaptic transmission. Furthermore, intrathecal injection of let-7b, at a higher dose (10 µg), induced persistent mechanical allodynia for >2 weeks, which was abolished in Tlr7-/- mice. The high dose of let-7b also induced microgliosis in the spinal cord. Of interest, intrathecal minocycline only inhibited let-7b-induced mechanical allodynia in male but not female mice. Our findings indicate that the secreted microRNA let-7b has the capacity to provoke pain through both neuronal and glial signaling, thereby establishing miRNA as an emerging neuromodulator.

SELECTION OF CITATIONS
SEARCH DETAIL