Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Sci Rep ; 10(1): 10133, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32576837

ABSTRACT

ß-hemoglobinopathies are caused by abnormal or absent production of hemoglobin in the blood due to mutations in the ß-globin gene (HBB). Imbalanced expression of adult hemoglobin (HbA) induces strong anemia in patients suffering from the disease. However, individuals with natural-occurring mutations in the HBB cluster or related genes, compensate this disparity through γ-globin expression and subsequent fetal hemoglobin (HbF) production. Several preclinical and clinical studies have been performed in order to induce HbF by knocking-down genes involved in HbF repression (KLF1 and BCL11A) or disrupting the binding sites of several transcription factors in the γ-globin gene (HBG1/2). In this study, we thoroughly compared the different CRISPR/Cas9 gene-disruption strategies by gene editing analysis and assessed their safety profile by RNA-seq and GUIDE-seq. All approaches reached therapeutic levels of HbF after gene editing and showed similar gene expression to the control sample, while no significant off-targets were detected by GUIDE-seq. Likewise, all three gene editing platforms were established in the GMP-grade CliniMACS Prodigy, achieving similar outcome to preclinical devices. Based on this gene editing comparative analysis, we concluded that BCL11A is the most clinically relevant approach while HBG1/2 could represent a promising alternative for the treatment of ß-hemoglobinopathies.


Subject(s)
Anemia, Sickle Cell/genetics , CRISPR-Cas Systems , Fetal Hemoglobin/genetics , Gene Editing/methods , Kruppel-Like Transcription Factors/genetics , Repressor Proteins/genetics , gamma-Globins/genetics , Anemia, Sickle Cell/therapy , Antigens, CD34 , Cells, Cultured , Gene Expression/genetics , Humans , Molecular Targeted Therapy , Mutation
2.
Bio Protoc ; 10(24): e3850, 2020 Dec 20.
Article in English | MEDLINE | ID: mdl-33855105

ABSTRACT

The mRNA therapeutics is a new class of medicine to treat many various diseases. However, in vitro transcribed (IVT) mRNA triggers immune responses due to recognition by human endosomal and cytoplasmic RNA sensors, but incorporation of modified nucleosides have been shown to reduce such responses. Therefore, an assay signifying important aspects of the human immune system is still required. Here, we present a simple ex vivo method called 'RNA ImmunoGenic Assay' to measure immunogenicity of IVT-mRNAs in human whole blood. Chemically modified and unmodified mRNA are complexed with a transfection reagent (TransIT), and co-incubated in human whole blood. Specific cytokines are measured (TNF-α, INF-α, INF-γ, IL-6 and IL-12p70) using ELISAs. The qPCR analysis is performed to reveal the activation of specific immune pathways. The RNA ImmunoGenic Assay provides a simple and fast method to detect donor specific - immune response against mRNA therapeutics. Graphic abstract: Schematic representation of RNA ImmunoGenic Assay.

3.
Mol Ther ; 27(4): 803-823, 2019 04 10.
Article in English | MEDLINE | ID: mdl-30905577

ABSTRACT

Protein supplementation therapy using in vitro-transcribed (IVT) mRNA for genetic diseases contains huge potential as a new class of therapy. From the early ages of synthetic mRNA discovery, a great number of studies showed the versatile use of IVT mRNA as a novel approach to supplement faulty or absent protein and also as a vaccine. Many modifications have been made to produce high expressions of mRNA causing less immunogenicity and more stability. Recent advancements in the in vivo lung delivery of mRNA complexed with various carriers encouraged the whole mRNA community to tackle various genetic lung diseases. This review gives a comprehensive overview of cells associated with various lung diseases and recent advancements in mRNA-based protein replacement therapy. This review also covers a brief summary of developments in mRNA modifications and nanocarriers toward clinical translation.


Subject(s)
Enzyme Replacement Therapy/methods , Lung Diseases/drug therapy , RNA, Messenger/administration & dosage , RNA, Messenger/genetics , Transcription, Genetic , Animals , Drug Delivery Systems/methods , Humans , Lipids/chemistry , Lung/metabolism , Lung Diseases/metabolism , Lung Diseases/pathology , Mice , Nanoparticles/chemistry , Polymers/chemistry , RNA Processing, Post-Transcriptional
4.
J Biomed Mater Res B Appl Biomater ; 107(6): 1824-1832, 2019 08.
Article in English | MEDLINE | ID: mdl-30466196

ABSTRACT

Membranes are an integral component of guided bone regeneration protocols. This pre-clinical study was aimed at enhancing the bioactivity of collagen membranes by incorporating plasmid DNA (pDNA) or chemically modified RNA (cmRNA) encoding bone morphogenetic protein-9 (BMP-9). In addition, we also endeavored to harness the regenerative potential of the periosteum by creating perforations in the membrane. Nanoplexes of polyethylenimine (PEI)-nucleic acids (PEI-pDNA or PEI-cmRNA encoding BMP-9) were incorporated into commercially obtained and perforated collagen membranes (PCM) to produce PCM-pDNA(BMP-9) or PCM-cmRNA(BMP-9). After structural characterization, the biodegradation kinetics of PCM, PCM-pDNA(BMP-9) and PCM-cmRNA(BMP-9) were assessed in simulated body fluid in vitro. Using a 24-well transwell plate system with bone marrow stromal cells (BMSCs) in the lower chamber and the PCM to be tested in the upper chamber, the in vitro bioactivity of different PCMs was evaluated by measuring various markers for osteogenesis in BMSCs. Alkaline phosphatase activity was assessed in BMSCs, after 7 and 11 days of exposure to PCM, PCM-pDNA(BMP-9), or PCM-cmRNA(BMP-9). Similarly, calcium deposition and Alizarin red staining in BMSCs were assessed after 14 days of exposure to the three different types of PCM. PCMs were then tested in vivo using the calvarial defect model in rats. After 4 weeks, animals were euthanized and bone specimens were harvested for micro-computed tomography and histological assessments. Incorporation of pDNA or cmRNA did not alter the biodegradation profile of PCMs. Alkaline phosphatase activity trended toward being higher in BMSCs exposed to PCM-cmRNA(BMP-9) or PCM-pDNA(BMP-9), when compared to BMSCs alone. Similar trends were observed when calcium deposition and alizarin red staining was evaluated. Calvarial bone defects treated with PCM-cmRNA(BMP-9) resulted in significantly higher bone volume/total volume % (BV/TV%), when compared to empty defects and trended toward being higher than defects treated with PCM-pDNA(BMP-9) and PCM alone. We demonstrate for the first time that resorbable PCM can be utilized to efficiently deliver pDNA and cmRNA of interest. The released pDNA and cmRNA encoding BMP-9 in this assessment was shown to be functional in vitro as well as in vivo. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B: 1824-1832, 2019.


Subject(s)
Bone Regeneration , Collagen , Membranes, Artificial , Skull , Animals , Bone Regeneration/drug effects , Collagen/chemistry , Collagen/pharmacology , DNA/chemistry , DNA/genetics , DNA/pharmacology , Growth Differentiation Factor 2/biosynthesis , Growth Differentiation Factor 2/genetics , Male , Plasmids/chemistry , Plasmids/pharmacology , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/pharmacology , Rats , Rats, Inbred F344 , Skull/injuries , Skull/metabolism , Skull/pathology
5.
Sci Rep ; 8(1): 16776, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30425265

ABSTRACT

Gene therapy has always been a promising therapeutic approach for Cystic Fibrosis (CF). However, numerous trials using DNA or viral vectors encoding the correct protein resulted in a general low efficacy. In the last years, chemically modified messenger RNA (cmRNA) has been proven to be a highly potent, pulmonary drug. Consequently, we first explored the expression, function and immunogenicity of human (h)CFTR encoded by cmRNAhCFTR in vitro and ex vivo, quantified the expression by flow cytometry, determined its function using a YFP based assay and checked the immune response in human whole blood. Similarly, we examined the function of cmRNAhCFTR in vivo after intratracheal (i.t.) or intravenous (i.v.) injection of the assembled cmRNAhCFTR together with Chitosan-coated PLGA (poly-D, L-lactide-co-glycolide 75:25 (Resomer RG 752 H)) nanoparticles (NPs) by FlexiVent. The amount of expression of human hCFTR encoded by cmRNAhCFTR was quantified by hCFTR ELISA, and cmRNAhCFTR values were assessed by RT-qPCR. Thereby, we observed a significant improvement of lung function, especially in regards to FEV0.1, suggesting NP-cmRNAhCFTR as promising therapeutic option for CF patients independent of their CFTR genotype.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/physiopathology , Cystic Fibrosis/therapy , Genetic Therapy/methods , Lung/physiopathology , Animals , Cell Line , Cystic Fibrosis/genetics , Disease Models, Animal , Humans , Maximal Expiratory Flow Rate/genetics , Mice , RNA, Messenger/chemistry , RNA, Messenger/genetics
6.
Mol Cell Pediatr ; 5(1): 9, 2018 Nov 14.
Article in English | MEDLINE | ID: mdl-30430274

ABSTRACT

BACKGROUND: ß-Thalassemia is an inherited hematological disorder caused by mutations in the human hemoglobin beta (HBB) gene that reduce or abrogate ß-globin expression. Although lentiviral-mediated expression of ß-globin and autologous transplantation is a promising therapeutic approach, the risk of insertional mutagenesis or low transgene expression is apparent. However, targeted gene correction of HBB mutations with programmable nucleases such as CRISPR/Cas9, TALENs, and ZFNs with non-viral repair templates ensures a higher safety profile and endogenous expression control. METHODS: We have compared three different gene-editing tools (CRISPR/Cas9, TALENs, and ZFNs) for their targeting efficiency of the HBB gene locus. As a proof of concept, we studied the personalized gene-correction therapy for a common ß-thalassemia splicing variant HBBIVS1-110 using Cas9 mRNA and several optimally designed single-stranded oligonucleotide (ssODN) donors in K562 and CD34+ hematopoietic stem cells (HSCs). RESULTS: Our results exhibited that indel frequency of CRISPR/Cas9 was superior to TALENs and ZFNs (P < 0.0001). Our designed sgRNA targeting the site of HBBIVS1-110 mutation showed indels in both K562 cells (up to 77%) and CD34+ hematopoietic stem cells-HSCs (up to 87%). The absolute quantification by next-generation sequencing showed that up to 8% site-specific insertion of the NheI tag was achieved using Cas9 mRNA and a chemically modified ssODN in CD34+ HSCs. CONCLUSION: Our approach provides guidance on non-viral gene correction in CD34+ HSCs using Cas9 mRNA and chemically modified ssODN. However, further optimization is needed to increase the homology directed repair (HDR) to attain a real clinical benefit for ß-thalassemia.

7.
Mol Ther Nucleic Acids ; 12: 530-542, 2018 Sep 07.
Article in English | MEDLINE | ID: mdl-30195789

ABSTRACT

The Cas9/guide RNA (Cas9/gRNA) system is commonly used for genome editing. mRNA expressing Cas9 can induce innate immune responses, reducing Cas9 expression. First-generation Cas9 mRNAs were modified with pseudouridine and 5-methylcytosine to reduce innate immune responses. We combined four approaches to produce more active, less immunogenic second-generation Cas9 mRNAs. First, we developed a novel co-transcriptional capping method yielding natural Cap 1. Second, we screened modified nucleotides in Cas9 mRNA to identify novel modifications that increase Cas9 activity. Third, we depleted the mRNA of uridines to improve mRNA activity. Lastly, we tested high-performance liquid chromatography (HPLC) purification to remove double-stranded RNAs. The activity of these mRNAs was tested in cell lines and primary human CD34+ cells. Cytokines were measured in whole blood and mice. These approaches yielded more active and less immunogenic mRNA. Uridine depletion (UD) most impacted insertion or deletion (indel) activity. Specifically, 5-methoxyuridine UD induced indel frequencies as high as 88% (average ± SD = 79% ± 11%) and elicited minimal immune responses without needing HPLC purification. Our work suggests that uridine-depleted Cas9 mRNA modified with 5-methoxyuridine (without HPLC purification) or pseudouridine may be optimal for the broad use of Cas9 both in vitro and in vivo.

9.
Cell Physiol Biochem ; 43(4): 1472-1486, 2017.
Article in English | MEDLINE | ID: mdl-29035889

ABSTRACT

BACKGROUND/AIMS: The Polo-like kinase 1 (Plk1) inhibitor volasertib is used in the treatment of malignancy. Volasertib is partially effective by triggering suicidal death or apoptosis of tumor cells. Similar to apoptosis of nucleated cells, erythrocytes may enter suicidal cell death or eryptosis, which is characterized by cell membrane scrambling with phosphatidylserine translocation to the cell surface and by cell shrinkage. Stimulators of eryptosis include energy depletion, hyperosmotic shock, oxidative stress and excessive increase of cytosolic Ca2+ activity ([Ca2+]i). The present study explored, whether volasertib impacts on eryptosis. METHODS: Human erythrocytes have been exposed to energy depletion (glucose withdrawal for 48 hours), hyperosmotic shock (addition of 550 mM sucrose for 6 hours), oxidative stress (addition of 0.3 mM tert-butylhydroperoxide [tBOOH] for 50 min) or Ca2+ ionophore ionomycin (1 µM for 60 min) in absence and presence of volasertib (0.5-1.5 µg/ml) and flow cytometry was employed to quantify phosphatidylserine exposure at the cell surface from annexin-V-binding, cell volume from forward scatter, [Ca2+]i from Fluo3 fluorescence, reactive oxygen species from 2',7'-dichlorodihydrofluorescein diacetate (DCFDA) fluorescence and ceramide abundance utilizing antibodies. For comparison, annexin-V-binding and forward scatter were determined following a 48 hours exposure of human leukemic K562 cells in RPMI-1640 medium to volasertib. RESULTS: Treatment with volasertib alone did not significantly modify annexin-V-binding or forward scatter in mature erythrocytes. Energy depletion, hyperosmotic shock, oxidative stress and ionomycin, all markedly and significantly increased the percentage of annexin-V-binding erythrocytes, and decreased the forward scatter. Volasertib significantly blunted the effect of energy depletion and hyperosmotic shock, but not of oxidative stress and ionomycin on annexin-V-binding. Volasertib did not significantly influence the effect of any maneuver on forward scatter. In K562 cells, volasertib enhanced annexin-V-binding and decreased the forward scatter. CONCLUSIONS: Volasertib is a novel inhibitor of erythrocyte cell membrane scrambling following energy depletion and hyperosmotic shock, effects contrasting the stimulation of K562 cell apoptosis.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Eryptosis/drug effects , Erythrocytes/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Pteridines/pharmacology , Cell Line, Tumor , Energy Metabolism/drug effects , Erythrocytes/cytology , Erythrocytes/metabolism , Glucose/metabolism , Humans , Osmotic Pressure/drug effects , Oxidative Stress/drug effects , Polo-Like Kinase 1
10.
PLoS One ; 12(8): e0183526, 2017.
Article in English | MEDLINE | ID: mdl-28846703

ABSTRACT

Cystic Fibrosis (CF) is the most common monogenic disease among people of Western European descent and caused by mutations in the CFTR gene. However, the disease severity is immensely variable even among patients with similar CFTR mutations due to the possible effect of 'modifier genes'. To identify genetic modifiers, we applied RNA-seq based transcriptomic analyses in CF patients with a mild and severe lung phenotype. Global gene expression and enrichment analyses revealed that genes of the type I interferon response and ribosomal stalk proteins are potential modifiers of CF related lung dysfunction. The results provide a new set of CF modifier genes with possible implications as new therapeutic targets for the treatment of CF.


Subject(s)
Cystic Fibrosis/genetics , Genotype , Interferon Type I/genetics , Ribosomal Proteins/genetics , Transcriptome , Adolescent , Adult , Child , Cystic Fibrosis/diagnosis , Female , Humans , Male , Mutation , Phenotype , Severity of Illness Index , Young Adult
11.
AAPS J ; 19(2): 438-446, 2017 03.
Article in English | MEDLINE | ID: mdl-28074350

ABSTRACT

Employing cost-effective biomaterials to deliver chemically modified ribonucleic acid (cmRNA) in a controlled manner addresses the high cost, safety concerns, and lower transfection efficiency that exist with protein and gene therapeutic approaches. By eliminating the need for nuclear entry, cmRNA therapeutics can potentially overcome the lower transfection efficiencies associated with non-viral gene delivery systems. Here, we investigated the osteogenic potential of cmRNA-encoding BMP-9, in comparison to cmRNA-encoding BMP-2. Polyethylenimine (PEI) was used as a vector to increase in vitro transfection efficacy. Complexes of PEI-cmRNA (encoding BMP-2 or BMP-9) were fabricated at an amine (N) to phosphate (P) ratio of 10 and characterized for transfection efficacy in vitro using human bone marrow stromal cells (BMSCs). The osteogenic potential of BMSCs treated with these complexes was determined by evaluating the expression of bone-specific genes as well as through the detection of bone matrix deposition. It was found that alkaline phosphatase (ALP) expression 3 days post transfection in the group treated with BMP-9-cmRNA was significantly higher than that in the group that received BMP-2-cmRNA treatment. Alizarin red staining and atomic absorption spectroscopy demonstrated enhanced osteogenic differentiation as evidenced by increased bone matrix production by the BMSCs treated with BMP-9-cmRNA when compared to cells treated with BMP-2-cmRNA. In vivo studies showed increased bone formation in calvarial defects treated with the BMP-9-cmRNA and BMP-2-cmRNA collagen scaffolds when compared to empty defects. The connectivity density of the regenerated bone was higher (2-fold-higher) in the group that received BMP-9-cmRNA compared to BMP-2-cmRNA. Together, these findings suggest that cmRNA-activated matrix encoding osteogenic molecules can provide a powerful strategy for bone regeneration with significant clinical translational potential.


Subject(s)
Bone Morphogenetic Protein 2/genetics , Bone Regeneration/genetics , Growth Differentiation Factor 2/genetics , Mesenchymal Stem Cells/cytology , Alkaline Phosphatase/genetics , Animals , Humans , Male , Osteogenesis/genetics , Polyethyleneimine/chemistry , RNA/administration & dosage , Rats, Inbred F344 , Transfection
12.
Gut ; 66(3): 473-486, 2017 03.
Article in English | MEDLINE | ID: mdl-27633923

ABSTRACT

OBJECTIVE: The generation of acinar and ductal cells from human pluripotent stem cells (PSCs) is a poorly studied process, although various diseases arise from this compartment. DESIGN: We designed a straightforward approach to direct human PSCs towards pancreatic organoids resembling acinar and ductal progeny. RESULTS: Extensive phenotyping of the organoids not only shows the appropriate marker profile but also ultrastructural, global gene expression and functional hallmarks of the human pancreas in the dish. Upon orthotopic transplantation into immunodeficient mice, these organoids form normal pancreatic ducts and acinar tissue resembling fetal human pancreas without evidence of tumour formation or transformation. Finally, we implemented this unique phenotyping tool as a model to study the pancreatic facets of cystic fibrosis (CF). For the first time, we provide evidence that in vitro, but also in our xenograft transplantation assay, pancreatic commitment occurs generally unhindered in CF. Importantly, cystic fibrosis transmembrane conductance regulator (CFTR) activation in mutated pancreatic organoids not only mirrors the CF phenotype in functional assays but also at a global expression level. We also conducted a scalable proof-of-concept screen in CF pancreatic organoids using a set of CFTR correctors and activators, and established an mRNA-mediated gene therapy approach in CF organoids. CONCLUSIONS: Taken together, our platform provides novel opportunities to model pancreatic disease and development, screen for disease-rescuing agents and to test therapeutic procedures.


Subject(s)
Cystic Fibrosis/therapy , Disease Models, Animal , Organoids/growth & development , Organoids/transplantation , Pancreas/cytology , RNA, Messenger/therapeutic use , Acinar Cells/cytology , Animals , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Gene Expression Profiling , Genetic Therapy , Humans , Mice , Organoids/cytology , Organoids/metabolism , Pancreas/growth & development , Pancreas/metabolism , Pancreatic Ducts/cytology , Phenotype , Pluripotent Stem Cells
13.
PLoS One ; 11(4): e0154001, 2016.
Article in English | MEDLINE | ID: mdl-27101288

ABSTRACT

Asthma is the most common chronic disease in childhood. Although several therapeutic options are currently available to control the symptoms, many drugs have significant side effects and asthma remains an incurable disease. Microbial exposure in early life reduces the risk of asthma and several studies have suggested protective effects of Toll-like receptor (TLR) activation. We showed previously that modified mRNA provides a safe and efficient therapeutic tool for in vivo gene supplementation. Since current asthma drugs do not take patient specific immune and TLR backgrounds into consideration, treatment with tailored mRNA could be an attractive approach to account for the patient's individual asthma phenotype. Therefore, we investigated the effect of a preventative treatment with combinations of Tlr1, Tlr2 and Tlr6 mRNA in a House Dust Mite-induced mouse model of asthma. We used chemically modified mRNA which is-in contrast to conventional viral vectors-non-integrating and highly efficient in gene transfer. In our study, we found that treatment with either Tlr1/2 mRNA or Tlr2/6 mRNA, but not Tlr2 mRNA alone, resulted in better lung function as well as reduced airway inflammation in vivo. The present results point to a potentially protective effect of TLR heterodimers in asthma pathogenesis.


Subject(s)
Asthma/therapy , Disease Models, Animal , RNA, Messenger/genetics , Toll-Like Receptors/genetics , Animals , Female , Mice , Mice, Inbred BALB C , Pyroglyphidae/pathogenicity
14.
Discov Med ; 21(113): 35-9, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26896600

ABSTRACT

Several tissue engineering strategies in the form of protein therapy, gene therapy, cell therapy, and their combinations are currently being explored for oral and craniofacial regeneration and repair. Though each of these approaches has advantages, they all have common inherent drawbacks of being expensive and raising safety concerns. Using RNA (encoding therapeutic protein) has several advantages that have the potential to overcome these limitations. Chemically modifying the RNA improves its stability and mitigates immunogenicity allowing for the potential of RNA to become an alternative to protein and gene based therapies. This brief review article focuses on the potential of RNA therapeutics in the treatment of disorders in the oral and craniofacial regions.


Subject(s)
Genetic Therapy/methods , Mouth Diseases/therapy , RNA/chemistry , RNA/therapeutic use , Regenerative Medicine/methods , Administration, Oral , Animals , Humans
15.
Mol Cell Pediatr ; 2(1): 11, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26589812

ABSTRACT

BACKGROUND: The immunogenicity and limited stability of conventional messenger RNA (mRNA) has traditionally restricted its potential therapeutic use. In 1992, the first clinical application of mRNA was reported as a potential protein-replacement therapy; however, subsequent investigations have not been made for almost two decades. Recent developments, including increased stability, controlling immunogenicity, as well as utilization of mRNA encoding zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and CRISPR-Cas9, have implicated modified mRNA as a very promising option for cancer immunotherapy, vaccines, protein expression replacement, and genome editing. This review aims to offer a summary of our present understanding of and improvements in mRNA-based drug technologies, along with a focus on the role in therapeutic options for pediatric respiratory diseases and hemoglobinopathies. CONCLUSIONS: This mini review summarizes the recent advances in modified mRNA-based therapy and its potential therapeutic effect in treating major pediatric diseases.

16.
J Control Release ; 218: 22-8, 2015 Nov 28.
Article in English | MEDLINE | ID: mdl-26415855

ABSTRACT

There exists a dire need for improved therapeutics to achieve predictable bone regeneration. Gene therapy using non-viral vectors that are safe and efficient at transfecting target cells is a promising approach to overcoming the drawbacks of protein delivery of growth factors. Here, we investigated the transfection efficiency, cytotoxicity, osteogenic potential and in vivo bone regenerative capacity of chemically modified ribonucleic acid (cmRNA) (encoding BMP-2) complexed with polyethylenimine (PEI) and made comparisons with PEI complexed with conventional plasmid DNA (encoding BMP-2). The polyplexes were fabricated at an amine (N) to phosphate (P) ratio of 10 and characterized for transfection efficiency using human bone marrow stromal cells (BMSCs). The osteogenic potential of BMSCs treated with these polyplexes was validated by determining the expression of bone-specific genes, osteocalcin and alkaline phosphatase as well as through the detection of bone matrix deposition. Using a calvarial bone defect model in rats, it was shown that PEI-cmRNA (encoding BMP-2)-activated matrices promoted significantly enhanced bone regeneration compared to PEI-plasmid DNA (BMP-2)-activated matrices. Our proof of concept study suggests that scaffolds loaded with non-viral vectors harboring cmRNA encoding osteogenic proteins may be a powerful tool for stimulating bone regeneration with significant potential for clinical translation.


Subject(s)
Bone Morphogenetic Protein 2/genetics , Bone Regeneration , Polyethyleneimine/chemistry , RNA/administration & dosage , RNA/chemistry , Alkaline Phosphatase/genetics , Animals , Bone Marrow Cells/cytology , Cell Survival , Craniocerebral Trauma/therapy , DNA/administration & dosage , DNA/chemistry , Genetic Therapy , Humans , Male , Mice, Inbred BALB C , Osteocalcin/genetics , Plasmids , RNA/pharmacology , RNA/therapeutic use , Rats, Inbred F344 , Stromal Cells/metabolism
18.
Am J Respir Cell Mol Biol ; 51(5): 709-20, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24828142

ABSTRACT

Whereas cigarette smoking remains the main risk factor for emphysema, recent studies in ß-epithelial Na(+) channel-transgenic (ßENaC-Tg) mice demonstrated that airway surface dehydration, a key pathophysiological mechanism in cystic fibrosis (CF), caused emphysema in the absence of cigarette smoke exposure. However, the underlying mechanisms remain unknown. The aim of this study was to elucidate mechanisms of emphysema formation triggered by airway surface dehydration. We therefore used expression profiling, genetic and pharmacological inhibition, Foerster resonance energy transfer (FRET)-based activity assays, and genetic association studies to identify and validate emphysema candidate genes in ßENaC-Tg mice and patients with CF. We identified matrix metalloproteinase 12 (Mmp12) as a highly up-regulated gene in lungs from ßENaC-Tg mice, and demonstrate that elevated Mmp12 expression was associated with progressive emphysema formation, which was reduced by genetic deletion and pharmacological inhibition of MMP12 in vivo. By using FRET reporters, we show that MMP12 activity was elevated on the surface of airway macrophages in bronchoalveolar lavage from ßENaC-Tg mice and patients with CF. Furthermore, we demonstrate that a functional polymorphism in MMP12 (rs2276109) was associated with severity of lung disease in CF. Our results suggest that MMP12 released by macrophages activated on dehydrated airway surfaces may play an important role in emphysema formation in the absence of cigarette smoke exposure, and may serve as a therapeutic target in CF and potentially other chronic lung diseases associated with airway mucus dehydration and obstruction.


Subject(s)
Airway Obstruction/immunology , Macrophage Activation/immunology , Macrophages, Alveolar/immunology , Matrix Metalloproteinase 12/immunology , Mucus/immunology , Pulmonary Emphysema/immunology , Airway Obstruction/metabolism , Animals , Bronchoalveolar Lavage Fluid/immunology , Cystic Fibrosis/genetics , Cystic Fibrosis/immunology , Cystic Fibrosis/metabolism , Dehydration/immunology , Dehydration/metabolism , Genomics , Macrophages, Alveolar/metabolism , Matrix Metalloproteinase 12/genetics , Matrix Metalloproteinase 12/metabolism , Mice, Knockout , Mucus/metabolism , Polymorphism, Single Nucleotide/genetics , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Emphysema/metabolism , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/immunology , STAT6 Transcription Factor/metabolism , Signal Transduction/immunology
19.
J Clin Invest ; 123(3): 1216-28, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23391720

ABSTRACT

Chemically modified mRNA is capable of inducing therapeutic levels of protein expression while circumventing the threat of genomic integration often associated with viral vectors. We utilized this novel therapeutic tool to express the regulatory T cell transcription factor, FOXP3, in a time- and site-specific fashion in murine lung, in order to prevent allergic asthma in vivo. We show that modified Foxp3 mRNA rebalanced pulmonary T helper cell responses and protected from allergen-induced tissue inflammation, airway hyperresponsiveness, and goblet cell metaplasia in 2 asthma models. This protection was conferred following delivery of modified mRNA either before or after the onset of allergen challenge, demonstrating its potential as both a preventive and a therapeutic agent. Mechanistically, FOXP3 induction controlled Th2 and Th17 inflammation by regulating innate immune cell recruitment through an IL-10-dependent pathway. The protective effects of FOXP3 could be reversed by depletion of IL-10 or administration of recombinant IL-17A or IL-23. Delivery of Foxp3 mRNA to sites of inflammation may offer a novel, safe therapeutic tool for the treatment of allergic asthma and other diseases driven by an imbalance in helper T cell responses.


Subject(s)
Asthma/prevention & control , Forkhead Transcription Factors/genetics , Interleukin-10/metabolism , RNA, Messenger/genetics , Airway Remodeling , Airway Resistance , Animals , Asthma/immunology , Asthma/metabolism , Cell Line , Cytidine/analogs & derivatives , Cytidine/chemistry , Female , Forkhead Transcription Factors/biosynthesis , Gene Expression , Genetic Therapy , Humans , Immunity, Innate , Inflammation Mediators/pharmacology , Inflammation Mediators/physiology , Interleukin-17/pharmacology , Interleukin-17/physiology , Interleukin-23/pharmacology , Interleukin-23/physiology , Mice , Mice, Inbred BALB C , Mice, Knockout , Pyroglyphidae/immunology , RNA, Messenger/chemistry , Th17 Cells/immunology , Th17 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Thiouridine/analogs & derivatives , Thiouridine/chemistry , Transfection
20.
J Immunol ; 190(3): 1276-84, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23277486

ABSTRACT

Pseudomonas aeruginosa persists in patients with cystic fibrosis (CF) and drives CF lung disease progression. P. aeruginosa potently activates the innate immune system, mainly mediated through pathogen-associated molecular patterns, such as flagellin. However, the host is unable to eradicate this flagellated bacterium efficiently. The underlying immunological mechanisms are incompletely understood. Myeloid-derived suppressor cells (MDSCs) are innate immune cells generated in cancer and proinflammatory microenvironments and are capable of suppressing T cell responses. We hypothesized that P. aeruginosa induces MDSCs to escape T cell immunity. In this article, we demonstrate that granulocytic MDSCs accumulate in CF patients chronically infected with P. aeruginosa and correlate with CF lung disease activity. Flagellated P. aeruginosa culture supernatants induced the generation of MDSCs, an effect that was 1) dose-dependently mimicked by purified flagellin protein, 2) significantly reduced using flagellin-deficient P. aeruginosa bacteria, and 3) corresponded to TLR5 expression on MDSCs in vitro and in vivo. Both purified flagellin and flagellated P. aeruginosa induced an MDSC phenotype distinct from that of the previously described MDSC-inducing cytokine GM-CSF, characterized by an upregulation of the chemokine receptor CXCR4 on the surface of MDSCs. Functionally, P. aeruginosa-infected CF patient ex vivo-isolated as well as flagellin or P. aeruginosa in vitro-generated MDSCs efficiently suppressed polyclonal T cell proliferation in a dose-dependent manner and modulated Th17 responses. These studies demonstrate that flagellin induces the generation of MDSCs and suggest that P. aeruginosa uses this mechanism to undermine T cell-mediated host defense in CF and other P. aeruginosa-associated chronic lung diseases.


Subject(s)
Cystic Fibrosis/complications , Flagellin/immunology , Immune Evasion/immunology , Immune Tolerance/immunology , Myeloid Cells/immunology , Pneumonia, Bacterial/immunology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/pathogenicity , Adolescent , Adult , Bacterial Proteins/genetics , Cells, Cultured/immunology , Culture Media, Conditioned/pharmacology , Cystic Fibrosis/microbiology , Disease Susceptibility , Female , Flagella/immunology , Flagella/physiology , Flagellin/genetics , Flagellin/pharmacology , Gene Expression Regulation/immunology , Humans , Immunity, Innate , Lung/microbiology , Male , Myeloid Cells/drug effects , Myelopoiesis/immunology , Pneumonia, Bacterial/etiology , Pneumonia, Bacterial/microbiology , Pseudomonas Infections/etiology , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/isolation & purification , Receptors, CXCR4/biosynthesis , Receptors, CXCR4/genetics , Receptors, CXCR4/immunology , T-Lymphocyte Subsets/immunology , Th17 Cells/immunology , Toll-Like Receptor 5/immunology , Up-Regulation/immunology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...