Skip to main content
Molecular Therapy logoLink to Molecular Therapy
. 2019 Mar 6;27(4):803–823. doi: 10.1016/j.ymthe.2019.02.019

Recent Developments in mRNA-Based Protein Supplementation Therapy to Target Lung Diseases

Itishri Sahu 1,2, AKM Ashiqul Haque 1,2, Brian Weidensee 1,2, Petra Weinmann 1, Michael SD Kormann 1,
PMCID: PMC6453549  PMID: 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.

Keywords: mRNA, lung, protein supplement, in vitro transcription, nanotransporter


mRNA has gathered attention as a new class of therapeutics for different diseases. Progress has been made in the field of protein replacement therapy by developing mRNA into a safe and efficient therapy. This article focuses on the delivery of mRNA therapeutics in the lung, and it provides an overview of lung architecture and associated diseases, such as asthma, cystic fibrosis, COPD, and SP-B deficiency.

Main Text

RNA, a fundamental molecule in the eukaryotic and prokaryotic cells and viruses, came recently into focus for therapeutic approaches:1, 2, 3 tRNAs for nonsense mutation correction, RNA aptamers for binding to a specific target molecule, and RNAi and long noncoding RNAs (lncRNAs) for gene regulation.4, 5, 6, 7, 8 mRNA gives rise to a new therapy for diseases associated with functional protein loss by supplementing the protein with a transcript encoding. The first study of in vitro-transcribed (IVT) mRNA in the late 1980s showed that this mRNA can be directly translated into a functional form immediately after transfection in vitro and in vivo.9, 10 This was shortly followed by a therapeutic application of mRNA in a temporary reversal of diabetes insipidus, unraveling its therapeutic potential.11

Therapeutic applications of mRNA are advantageous due to its unique properties. Cytoplasm being mRNA’s functional site requires no transportation across the nuclear membrane. Furthermore, due to the transient nature and biodegradability of mRNA, permanent adverse effects can potentially be avoided.12 This includes preventing the permanent manipulation of the genome, making mRNA a prevalent molecule for protein supplementation therapy.13, 14, 15 Several aspects of mRNA have to be addressed in order to achieve therapeutic benefits: potential immunogenicity mediated by innate immune system reactions (pattern recognition receptor),16, 17, 18, 19 degradation of single-stranded mRNA by nucleases, and its negative charge that inhibits the passive crossing of the cell membrane. Concerning these obstacles, solutions such as chemical modifications of nucleosides to reduce immunogenicity and usage of nanocarriers to facilitate crossing the cell membrane are emerging. The recent advancements in the field of nanocarriers suggest the possibility to customize particles for target organs.20, 21

Based on our research interest in lungs, this review focuses on protein replacement therapy of lung disease, especially monogenetic diseases, such as cystic fibrosis (CF) and surfactant protein B (SP-B) deficiency, as well as multifactorial diseases, such as chronic obstructive pulmonary disease (COPD) and asthma. To achieve targeted therapy options, lung structure and methods to reach specific lung cell populations are critical. Therefore, this review gives an overview of lung cell populations and diseases associated with them. Furthermore, a comprehensive summary of mRNA transcript improvements by chemical modification of nucleosides or capping and nanocarriers to target lung cell populations is also featured.

Lung Architecture and Disease Pathology

The lung has a very unique architecture to enable efficient transfer of oxygen and carbon dioxide required for oxidative metabolism. Inhaled gases travel through the airway tubes via trachea bronchi and bronchioles to the alveoli enriched with blood vessels, the primary site of gas exchange. Inflation and deflation of the lung is a prerequisite for gas exchange at the alveoli. This process requires multiple components like the extracellular matrix, smooth muscle cells, and cartilage for support and flexible collagen and the elastin fiber network for flexibility during inflation and deflation. Precisely regulated surface fluids, electrolytes, and mechanical activity of secretory and ciliated cells determine the mucociliary clearance, whereas on the other hand the epithelium maintains the barrier function.

The airway epithelium (tracheal and bronchiolar) consists of goblet cells, brush or tuft cells, ciliated cells, basal cells, neuroendocrine or neuroepithelial bodies, club cells, lineage-negative epithelial progenitor (LNEP) cells, and the newly identified ionocytes.22, 23 The ducts of submucosal glands consist of goblet cells, serous cells, and myoepithelial cells. The alveolar epithelium consists primarily of alveolar type I (ATI) cells (∼95%), macrophages, and alveolar type II (ATII) cells, with close association to capillary endothelial cells of pulmonary microcirculation.24 The terminal airway ducts and alveoli are supported by fibroblasts and myofibroblasts producing extensive elastin-collagen networks, which help with inflation and deflation.25, 26, 27 Other important cell populations include stem cells and immune cells, which help in region-specific regeneration and protection against pathogens, respectively. Abnormalities of lung mechanics are observed in various diseases, including CF,28, 29 asthma,30, 31 idiopathic pulmonary fibrosis (IPF),32, 33 COPD,34, 35 and bronchiolitis obliterans.34, 36 These conditions are detrimental in nature and involve multiple factors, such as increased resistance of lung tissue due to fibrosis of the lung; collapsing tubes or thickening of airway walls due to mucus overproduction; and the loss of ciliary function, resulting in airflow obstruction, mucus plugging, chronic infection, and inflammatory damages. In that process, multiple cell populations undergo remodeling, directly contributing to clinical symptoms.

Broncho-Epithelial Cells and Associated Diseases

Goblet cells, present in the broncho-epithelia and submucosal glands, produce mucins (MUC5B and MUC5AC), and they are differentiated from basal and club cells upon various stimuli (toxic substances, pathogens, particles, and neural and innate immune signals). Goblet cells are not only helping to establish the innate immune system but also key players in pulmonary diseases, as described before.37 These cells produce cytokines and chemokines that recruit and educate innate immune cells, including dendritic cells (DCs), innate lymphoid cells, and eosinophils. This contributes to the CD4+ T helper cell 2 (Th2)-mediated immune response typical of asthma.38, 39 Hyper-production of mucus and goblet cell metaplasia are characteristics of Th2-mediated and non-mediated inflammation (pathogen mediated), which leads to complications in CF, COPD, and IPF.40, 41 The fluids for hydration are regulated by serous cells that line the acinar region of submucosal glands and are important for the pathology of CF. The mucins are precisely balanced by fluid, and electrolyte transport enables rapid secretion and dispersal of mucus onto the airway surfaces and the movement of the mucus gel up the airway by ciliary activity. In CF, the secretion of chloride and bicarbonate is impaired, disrupting mucociliary clearance due to mutations in CF transmembrane conductance regulator gene (CFTR), which leads to recurrent airway infections, sinusitis, bronchiectasis, and pulmonary tissue remodeling.42

Brush cells, containing distinctive apical microvilli, are found in multiple organs, e.g., pancreas, intestine, nose, and trachea. Brush cells are known to play an important role in activating the innate immune system in the intestine43 and nose (Tas2R receptors),44 however, a similar phenomenon in airway trachea requires testing. Recent studies in the trachea have indicated their role as chemosensory for immune surveillance and as respiratory regulators.45 These might be responsible in transducing signals regulating wheezing and coughing during episodes of asthma. However, further studies are required to understand their role.46

Ciliated cells are characterized by their multiple apical, motile cilia composed of structural proteins and motor proteins (dynein)47, 48, 49 that regulate the coordinated bidirectional beating critical for particle and pathogen clearance.50, 51 Ciliated cells respond to both physical52 and chemical53 stimulation. Mucociliary clearance can be affected by ciliary dysfunction, impaired fluid secretion, disruption of epithelial cell lining, or lack of cough. This impairment can initiate an inflammatory response, damaging the airway epithelium. Disruption in ciliated cell function results in recurrent and persistent infections, morbidity, and mortality in chronic pulmonary disorders.54 In COPD, direct evidence has been provided of suppressed ciliary beating in nasal epithelium55 with normal mucus production. Cigarette smoking has been shown to have a detrimental effect on the number and size of cilia in vitro,56 whereas in vivo a slight increase in ciliary beating initially followed by significant loss of cilia over prolonged time was observed.57 Primary ciliary dyskinesia (PCD) resulting from ciliary dysfunction is caused by recessive mutations in one of multiple genes involved at different points in cilium structure, assembly, and function, which include DNAI,58 DNAH(5,59 1160), ARMC4,61 TXNDC3,62 HEATR2,63 HYDIN,64 CDC (39,65 40,66 65,67 103,68 114,69 15170), DNAAF(1,71 2,72 373), RHSP(4A, 9),74 DYX1C1,75 LRRC6,76 ZMYND10,77 CCNO,78 and recently identified MCIDAS.79

Club cells are columnar, secretory cells that express high levels of cytochrome P450-detoxifying enzyme CYP2F, surfactant proteins (SP-A, SP-B, and SP-D), and innate immune proteins, including defensins, lactoferrin, and secretaglobins (SCGB1A1 and SCGB3A1).80 Upon stimulus or injury, these differentiate into alveolar cells,81, 82 goblet cells,38, 83, 84 or ciliated cells.85 However, in humans, these are only localized in terminal bronchioles, and they may play a role in the maintenance of distal bronchioles.86 Club cells being progenitor cells also plays an important role in the repair mechanism of the airway epithelium, and, therefore, they are connected to damage responses in CF, COPD, and IPF.80 Club cells also metabolize chemical toxins (e.g., naphthalene) to toxic compounds that selectively kill club cell subsets. SP-A and SP-D (C-type lectins) are responsible for host defense, enhancing the clearance of various microbial pathogens, whereas the secretory lipids and proteins (SP-B and SP-C) help in minimizing surface tension and collapsing forces caused by inhaled gases when in direct contact with alveolar structures, protecting peripheral saccules from atelectasis during ventilator cycles.87

Neuroendocrine cells represent less than 1% of the airway epithelium,88 and they are found either isolated or in clusters known as neuroepithelial bodies located at precise airway branch points.89 The localization aids in their role as airway (environmental) sensors (acidosis, hypoxia, and hypercarbia),90, 91 and clustering is required for the appropriate innate immune responses.92 The response to a stimulus is via the release of stored amines (serotonin) and peptides (calcitonin gene-related peptide [CGRP]).92 Hyperplasia is associated with a wide range of congenital and infantile lung disorders. Though the underlying cause is unknown, NKX2-1 mutation has been associated with neuroendocrine hyperplasia of infancy (NEHI).93

Ionocytes (Foxi1+) are the rare cell type of the airway epithelium recently characterized, in both mice and humans, by two independent research groups.22, 23 These reside at multiple levels of the airway tree, and they are needed to maintain airway surface physiology, including mucus viscosity.23 Foxi1 is already known to regulate V-ATPase, which is important for transport and fluid pH in other cell types in skin.94, 95 Knockout in a mouse model reduced Cftr and Ascl3 expression, indicating the role of Foxi1 in CFTR regulation.23 Montoro et al.23 also performed pulse-sequence tracing that indicated the basal cell lineage, with an increased expression of CFTR.

Basal cells are progenitor cells96, 97, 98 that are regulated by NOTCH signaling to give rise to ciliated cells (NOTCH), goblet cells (NOTCH+), and club cells (NOTCH2+).99, 100, 101, 102 Due to their basal proximity, these cells interact with the columnar epithelium; underlying mesenchymal cells; basal membrane; neurons; and also lymphocytes, inflammatory cells, and DCs.103 A loss in tight regulation of basal cell differentiation can result in inappropriate cell fate determination, leading to pathological airway remodeling. This includes epithelial hypoplasia (proliferation failure), basal cell hyperplasia (excessive proliferation with no differentiation), goblet cell metaplasia or hyperplasia instead of ciliated cell generation, and squamous metaplasia (suprabasal cells) instead of luminal cells. Pathological airway remodeling occurs frequently in association with CF, COPD, and chronic asthma.103 Araya et al.104 in their study showed that hyper-proliferating basal cells secrete cytokines (interleukin-1β [IL-1β]) that promote airway wall fibrosis via transforming growth factor β (TGF-β) signaling in COPD.

Alveolar Cells and Associated Diseases

Alveolar type I cells (ATI) are squamous cells lining the alveolar compartment involved in gas exchange. These are terminally differentiated cells (lifespan ∼120 days) that form a barrier to sense microbial products and generate inflammatory responses.105, 106 These cells undergo excessive physical and chemical stresses due to their higher exposure, and they require constant regulated repair.107, 108 A defect in repair directly and indirectly contributes toward injurious manifestations of the lung, leading to diseases like acute respiratory distress syndrome (ARDS) and IPF.109, 110 ATII (progenitor of ATI cells) are cuboidal cells and cover about 7% of the total alveolar surface. These produce surfactant lipids (phosphatidylcholine) and surfactant proteins (SP-A to -D). Congenital SP-B deficiency leads to death87, 111 soon after birth; however, targeted disruption of SP-C,112 SP-A,113 and SP-D114, 115 gene loci does not show detrimental effects. In cases with acute respiratory distress syndrome, a decrease in the expression of SP-B is also observed.116, 117

Stem Cells of the Airways

To maintain the constant dynamic function of the lung, it is very crucial that the respiratory epithelium is equipped with fast and extensive regenerative ability following injury. Airway basal cells and ATII cells have been known for their role in repair of the airway epithelium. Recently, studies have identified distinct niches throughout the lung that can mediate graded and region-specific responses.118, 119 Myoepithelial cells (MECs) and bronchoalveolar stem cells (BASCs) are a couple of the established stem cells in lungs. It was well established that the innervated MECs encircle the submucosal glands and mediate mucus secretion in response to neural inputs, which can activate massive secretory responses after stimulation by irritants and toxins (also reviewed in Boers et al.86 and Yei et al.120).37, 42, 121 However, recent work by Lynch et al.122 and Tata et al.123 have explained the potency of MECs in generating seven cell types of surface airway epithelium and the submucosal gland following airway injury. These can be activated via Sox9 or Lef-1 transcriptional signaling, and they can be exploited for regenerative medicine. BASCs have been identified as stem cells that co-express both club cell and ATII cell markers.124 BASCs sorted by flow cytometry and cultured in vitro showed differentiation, self-renewability, and response to injury.124, 125, 126 Lineage-tracing studies have revealed the BASC’s ability to give rise to alveolar epithelial cells in vivo127, 128 and its contribution to homeostasis and repair, along with club and ATII cells.127, 128, 129, 130

Respiratory Diseases Targeted by Protein Replacement Therapy

SP-B Deficiency

SP-B deficiency is a rare genetic disease leading to neonatal lethality, including interstitial lung disease (ILD) and ARDS.131, 132, 133, 134, 135 SP-B is crucial for breathing transition of neonates at birth, and it helps in reducing surface tension of the alveolus. Dipalmitoylphosphatidylcholine (DPPC) is the principal surface tension-reducing component that combines with hydrophobic SP-B or SP-C peptides to form stable surfactant film.136 Surfactant supports rapid adsorption and insertion of phospholipids, reduction in surface tension upon compression, and rapid re-spreading during expansion. Changes in surfactant lead to alveolocapillary leakage, alveolar instability, compromised gas exchange, and respiratory failure. Both SP-B and SP-C peptides are processed from their pro-peptide forms to their functional form. Absence of proSP-C processing leads to the accumulation of misprocessed SP-C, consisting of the mature peptide with an N-terminal extension (relative molecular weight [Mr] ∼6,000), and to a significant decrease in mature SP-C peptide in alveolar surfactant. ProSP-C processing is also closely related to SP-B expression.87, 111, 137 The combinatorial effect exacerbates lung function at birth, and in vivo studies showed respiratory failure in selective loss of SP-B in adult.138

Natural surfactant replacement, such as Survanta beractant (modified bovine surfactant 8 mg SP-B/mL) by Abbott Laboratories, Curosurf (porcine surfactant 80 mg/mL) by Chiesi, and Infasurf (calf surfactant), are a few of the FDA-approved preventive medications for infants with ARDS or premature babies at the risk of developing RDS. Accompanied by physical measures, corticosteroids, or immunosuppressants and repeated lung lavage, surfactant replacements have shown improvement in the disease condition until lung transplantation.139, 140, 141, 142, 143, 144 The potential risks with animal-derived protein include immunological reactions and transmission of animal-derived diseases, justifying the need for standardized human-like alternatives.145, 146, 147 One alternative involves synthetic mimics that have shown superior surfactant properties.144, 148

Since SP-B deficiency is a monogenetic disorder, it acts as a perfect model for gene therapy. Both DNA- (virus120 and plasmid149) and mRNA-150 based gene supplementation have been tested in a conditional SP-B-knockout mouse model, which indicated improvements in lung function and SP-B expression and a significant increase in survival. Kormann et al.150 showed for the first time that intratracheal (i.t.) instillation of modified SP-B mRNA to the lung can restore up to 71% of the wild-type SP-B expression, and the treated conditional SP-B-knockout mouse model survived until the predetermined end of the study of 28 days. Presently, the Rudolph team from Ethris holds a patent for pulmonary delivery of mRNA with polyethylenimine (PEI) (US patent application 20150126589), and their teaming up with AstraZeneca and MedImmune could bring the therapy closer to the reach of patients.151

Asthma

Asthma is a multifactorial disease and can be characterized by airway obstruction, chest tightness, wheezing, cough, and breathlessness, followed by recurrent pneumonia or bronchitis. The initiating event in asthmatic airway diseases revolves around interactions between DCs and T cells. DC and T cell interactions favor the generation of Th2, leading to eosinophilia, mucus hypersecretion, and chronic airway inflammation.152, 153 The overactive Th2 response induces the production of cytokines and chemokines, followed by a cascade of immune-activating events, leading to changes in airway smooth muscle contractility,154 a characteristic of asthma. Studies by Hellings et al.155 and Wilson et al.156 showed that Th17 cells orchestrate airway inflammation by enhancing neutrophil recruitment to the lung. The Th2-mediated immune response can be contained via Th1-type cytokines (by Th1 cells), IL-10, and TGF-β (by T regulatory cells [Tregs]), but the roles of IL-17 and IL-22 (by Th17 cells) are debated.155, 157, 158 Both circulatory and airway fluids of asthmatic patients indicate increased IL-17 levels155 and decreased airway Tregs,159 indicating an imbalance in Th2 regulation.

Corticosteroid treatment is found to suppress the Th2 immune response via increased Foxp3+ Tregs in asthmatic patients.160 Similar results were found with exposure to microbes161, 162 influencing Treg expression, modulation of IL-6,163 prostanoids,164 and tumor necrosis factor (TNF) pathway enhancement.165 Mays et al.166 has successfully demonstrated the protective role of Foxp3 by delivering chemically modified Foxp3 mRNA into the lung of an asthma mouse model. Site-specific instillation of chemically modified Foxp3 mRNA can modulate both Th2 and Th17 responses in an IL-10-dependent manner.166 Local administration of Foxp3 mRNA can influence the balance among Treg, Th2, and Th17 responses, and it can reduce side effects in terms of the anti-tumoral and anti-infective167, 168 effects of Tregs in comparison to systemic delivery.165 Kormann et al.169 produced a unique insight into Toll-like receptors (TLRs), as polymorphisms in TLRs 1, 6, and 10 have shown protective effects on atopic asthma in humans by forming heterodimers with TLR2. A subsequent study by Zeyer et al.170 demonstrated that Tlr1/2 and Tlr2/6 mRNA instillation in the lungs of a house dust mite-induced mouse model of asthma reduced airway resistance by 40%.

CF

CF, caused by mutations in the CFTR gene, is the most common life-limiting autosomal-recessive disease in the Caucasian population, and it affects more than 80,000 people worldwide. Around 2,000 mutations have been identified and are categorized into 6 classes, ranging in severity from no production of functional protein to decreased synthesis, stability, or function of CFTR protein. CFTR protein acts as a small conductance ATP and cyclic AMP (cAMP)-dependent chloride channel, found at the apical side of epithelial cell lining of most exocrine glands. In the lung epithelium (ionocytes and ciliated and goblet cells), CFTR ensures the secretion of chloride ions, resulting in more hydration and regulated mucus clearance in the airway. A lack of functional CFTR leads to decreased chloride secretion and increased sodium absorption, resulting in dry and rigid mucus production by goblet cells.171 An increase in inflammatory response is also observed, possibly due to the breakdown of elastin fibers by neutrophil elastase and reduced IL-10.172 The defective mucus clearance enables further pathogen (Pseudomonas aeruginosa and Staphylococcus aureus) colonization, repetitive inflammatory responses that result in irreparable lung damage, and ultimately cardiac arrest. Defects of the CFTR channel lead to a failure in LPS recognition, endocytosis of pathogens, and changes in airway fluid composition. This inactivates beta defensins, causing detrimental effects on the primary defense in the lung.

A readthrough agent for CFTR class I mutation (Ataluren, PTC Therapeutics) showed potential benefits in vivo by increasing CFTR production and function;173 however, it failed phase III clinical trial due to a lack of efficacy.174 Channel modulators categorized into potentiators, correctors, and amplifiers have been used for CF treatment. Potentiators (ivacaftor, Kalydeco) can help in gating and conduction mutations by increasing the open probability of the CFTR channels. Correctors (tezacaftor and lumacaftor) improve CFTR trafficking by facilitating the formation of correct 3D protein structure. Combinations of potentiator and corrector (Orkambi and Symdeko, Vertex Pharmaceuticals) are commercially available only for patients with F508del mutation, expanding the modulators also for application on class II mutations.175, 176 A triple combination of two correctors (VX-659, tezacaftor) and one potentiator (ivacaftor) has also shown greater potency in pre-clinical trials. PTI-428 (Proteostasis Therapeutics), an amplifier to increase the amount of CFTR protein, is in phase II trial. Eluforsen (QR-010) can bind to defective CFTR RNA, and it has shown increased CFTR function by nasal potential difference test in compound heterozygous or homozygous F508del CF patients.177

Protein replacement therapy with DNA, protein, or mRNA holds a great potential as a universal therapy for curing the underlying defect of CF. Initial attempts of in vivo protein transfer via phospholipid liposomes into the apical membrane of nasal epithelia of CFTR-knockout mice showed limited membrane incorporation by electron microscopy but improvement of nasal potential difference (NPD) measurement.178 Similarly, DNA-based vectors (viral and plasmid) were tested by Alton’s group (pGM169/GL67A), reaching phase II clinical trials with modest improvement in FEV1 after repeated administration but no improvement in patient’s quality of life.179, 180 Haque et al.20 have observed a significant improvement in CFTR protein translation, expression, and function in vitro (CFBE41o- and 16HBE14o-) and in vivo (CFTR-knockout mice) by administering chemically modified human CFTR (hCFTR) mRNAs complexed with chitosan-coated poly(lactic-co-glycolic acid) (PLGA) nanoparticles. The study also showed a substantial improvement in FEV0.1 up to 89% of the level of a healthy control group. Airway compliance and resistance are also significantly improved by the treatment with chemically modified hCFTR mRNAs. A significant decrease in chloride concentration (around 50%) was also observed, indicating a restoration of CFTR in the duct compartment of submucosal glands and thus leading to improved chloride absorption.20 A separate study from Robinson et al.181 confirmed nasal application of chemically modified CFTR mRNA can recover up to 55% of the net chloride efflux characteristic of healthy mice. Bangel-Ruland et al.182 demonstrate restoration of cAMP-induced CFTR current following transfection of CFBE41o- cells with wild-type CFTR-mRNA similar to the values seen in 16HBE14o- control cells. Translate Bio is also working on CFTR-encoding mRNA (MRT5005) and has entered phase I/II clinical trial (Table 1). All these studies prove the potential of mRNA as a promising therapeutic in CF patients, irrespective of their CFTR mutation status.

Table 1.

Current Clinical Trials Involving mRNA Delivery

Name Disease Genetic/Protein Target Administration Route Administration Vehicle ClinicalTrials.gov Identifier Phase
Lipo-MERIT melanoma tumor-associated antigens intravenous infusion mRNA-Lipoplex NCT02410733 1
TNBC-MERIT triple-negative breast cancer tumor-associated antigens intravenous infusion mRNA-Lipoplex NCT02316457 1
IVAC mutanome/warehouse triple-negative breast cancer patient-specific antigens intra-nodal naked NCT02035956 1
mRNA-1851 influenza A Hemagglutinin 7 (H7) protein intramascular injection not disclosed Not disclosed 1
mRNA 1440 influenza A Hemagglutinin 7 (H7) protein intramuscular injection not disclosed not disclosed 1
CV7201 rabies rabies virus glycoprotein intramuscular injection naked NCT02241135 1
CV8102 HIV, rabies, RSV RNA-based adjuvant intramuscular injection naked NCT02238756 1
mRNA MRK-1777 not disclosed vaccine intradermal not disclosed not disclosed 1
mRNA AZD-8601 cardiovascular diseases VEGF-A intramuscular injection naked NCT02935712 1
mRNA-1325 Zika viral antigenic protein intramuscular injection lipid nanoparticle NCT03014089 1/2
CV9103 prostate cancer tumor-Specific antigen autologous dendritic cell therapy naked NCT01197625 NCT00831467 1/2
MRT5005 cystic fibrosis CFTR nebulization to the respiratory tract lipid nanoparticle NCT03375047 1/2
AGS-004 HIV vaccine autologous dendritic cell therapy naked NCT01069809, NCT02707900 1/2
AGS-003-LNG non-small-cell lung cancer tumor-specific antigen autologous dendritic cell therapy naked NCT02662634 2
iHIVARNA-01 HIV HIV-target antigen intranodal route naked NCT02888756 2
AGS-003 renal cell carcinoma tumor-specific antigen autologous dendritic cell therapy naked NCT01482949 NCT00678119 NCT01582672 2/3

Modified from Kaczmarek et al.321

COPD and COPD-like Symptoms in α1-Antitrypsin Deficiency

COPD is a progressive and largely irreversible smoking-related disease characterized by small airway obstruction, emphysema, and chronic bronchitis. It is mainly attributed to long-term exposure to tobacco, toxic gases, and particles, activating both innate and adaptive immune responses. The innate immune defense includes tight junctions, TLRs of epithelial barrier, macrophages, and alveolar fluid (secreted by ATII cells of lung). A second line of defense includes exudation of plasma and circulating effector cells into damaged tissue, regulated by IL-1, IL-8, and TNF-α. The infiltration of both bronchial and alveolar tissue with macrophages, B and T lymphocytes, and eosinophils has been associated with emphysematous destruction. Both the responses are linked to tissue repair and remodeling that increase mucus content of airway lumen and metaplasia of mucous and squamous cells. This leads to a thickened wall and narrowed lumen of conducting airways. Second, emphysema limits air flow by reducing elastic recoil pressure for exhaling air during forced expiration.183

α1-antitrypsin deficiency (AATD) can cause COPD-associated symptoms like emphysematous destruction along with innate inflammation in lung due to an imbalance in protease and antiprotease homeostasis.184 In lungs, AAT has the major physiological function of protecting the healthy but fragile alveolar tissue from proteolytic damage of neutrophil elastases.185 The AATD is largely associated with mutations within SERPINA1, resulting in abnormal protein folding, intracellular retention, and consequently low serum levels.186 However, multiple other factors also contribute to disease severity, and much research is being done to obtain a comprehensive picture for enabling better diagnosis.187, 188, 189, 190, 191, 192, 193, 194, 195, 196 Danozol was found to significantly improve AAT circulating levels197 without eliciting side effects. Another approach is to inhibit polymerization of AAT by small molecules,198 peptides,199 autophagy-enhancing drugs200, 201 (ClinicalTrials.gov: NCT01379469 Tregretol phase II clinical trial, rapamycin202), and RNA silencing of mutant AAT in liver hepatocytes.203, 204, 205 Intravenous (i.v.) augmentation of plasma-derived AAT (Bayer Biologicals, ZLB Behring, Baxter Healthcare)206, 207 is an established method to raise circulating levels of AAT in blood and bronchoalveolar lavage fluid (BALF), slowing the progression of lung destruction.208

Various routes of administration and vectors have been tested for gene delivery of AAT in various animal models (rat, mouse, and dog), with varying expression efficiency in terms of time and localization (reviewed in detail209). However, only AAVrh.10hAAT (Adverum Biotechnologies, ClinicalTrials.gov: NCT02168686) has entered the phase I/II clinical trial to assess the safety and therapeutic level expression of M1-type AAT in the serum and alveolar epithelial lining fluid.210 Connolly et al.21 have shown successful expression of AAT with liposome-encapsulated SerpinA1 mRNA in vitro and in vivo after transfection. In an independent study, Michel et al.211 observed significant expression of AAT in vitro and ex vivo along with a significant reduction in elastase activity.

Steps toward Pharmacologically Auspicious mRNA

In recent years substantial efforts have been made for engineering mRNA with diverse pharmacokinetic properties (Figure 1). Modifications of structural elements such as 5′ Cap, 5′ and 3′ UTRs, poly(A) tail, and the coding region were the main focus.14, 212, 213, 214, 215

Figure 1.

Figure 1

mRNA IVT, Modifications, and Function and Timeline

Overview of milestones in protein supplementation therapy, in vitro transcription, and mRNA modification. White boxes, important milestones for the development of mRNA therapy;250 blue boxes, evolution of different cap structures;215, 220, 322 red, green, and gray boxes, 5′ UTR, 3′ UTR, and poly(A) tail, respectively, the addition of regulatory elements in the modification of mRNA;14, 312, 313 yellow boxes, nucleoside modifications and sequence optimizations in the development for mRNA therapy.215, 230, 238, 240, 314, 317

5′-Capping for Stability and Immune Evasion

5′-capping is vital for the robust translation of mRNA as the natural 7-methylguanosine (m7G/Cap0), and it is connected by a 5′-to-5′ triphosphate bridge to the first nucleotide. Translation is initiated by binding to eukaryotic translation initiation factors (eIF4E and eIF4G), and mRNA deterioration is controlled by binding with Dcp1, Dcp2, and DcpS (mRNA-decaying enzymes).57, 216, 217, 218 In in vitro transcription, m7G possesses the risk of constructing uncapped or inactive IVT mRNA, as the m7G and GTP compete for incorporation. m7GpppG cap was the first step to circumvent the restriction of m7G, however, substantial proportions of m7GpppG analog were incorporated in the reverse direction and thus yielded substandard translational activity.9, 219 Anti-reverse cap analog (ARCA; m27,3′-OGpppG) can counteract the reverse integration and skip degeneration by Dcp2, and, thus, it results in superior translational efficiency and extended half-life.13, 150, 220 Study on viral capping systems reveals that the 2′ ribose position of the first cap-proximal nucleotide is 2′O-methylated to form a Cap 1 structure (m7GpppN2′Om N), and, in ∼50% of transcripts, the second cap-proximal nucleotide is 2′O-methylated to form a Cap 2 (m7GpppN2′OmN2′Om) structure.221 Cap 1 2′O-methylation has been described to reduce recognition by pattern recognition receptors (e.g., interferon [IFN]-induced protein with tetratricopeptide repeats-1 and 5 [IFIT1 and IFIT5] and retinoic acid-inducible gene I [RIG-I]) in comparison to Cap 0.222, 223, 224

Modification in UTRs

The poly(A) tail plays an important role in regulating the stability and translational efficiency (half-life) of mRNA by preventing deadenylation by poly-specific nucleases.225 Integration of the poly(A) tail during IVT mRNA synthesis can be conducted by encoding the poly(A) stretch in the template or by a two-step enzymatic reaction using recombinant poly(A) polymerase.226 The ideal length of the poly(A) tail is between 120 and 150 nt, and the 3′ end of the poly(A) tail should not be concealed by additional bases.14, 213 5′ and 3′ UTRs also play a vital role in the stability and expression of IVT mRNA by harboring several sequence elements. For example, mRNAs with adenosine in 5′ UTR can form a complex with Lsm1-7 at both the 5′ and 3′ ends and circularize the transcript to inhibit degradation by exosome and Dcp1/2.227 Adenine uracil (AU)-rich elements in the 3′ UTR can destabilize the mRNA, and they might provide a mechanism to limit the duration of protein production.228

Post-transcriptional Modifications

Post-transcriptional chemical modifications of RNA are not uncommon, and over 100 modifications are listed by different studies.229 In mRNA, only a small subset of these naturally occurring modifications is reported to be essential for reducing innate immune response and improving mRNA expression and stability.215, 230, 231 N6-methyladenosine (m6A) is one of the most frequent modifications in eukaryotic mRNA. Insulin-like growth factor 2 (IGF2) mRNA-binding proteins 1, 2, and 3 (IGF2BP1/2/3) preferentially recognize m6A mRNA and guard the modified mRNA against decay.232 Based on the studies of Kormann and Warren et al.,233 the first generation of modified mRNAs containing 5-methylcytidine (m5C) or pseudouridine (Ψ-UTP) reduces innate immune responses and enhances translation. TLR3, TLR7, TLR8, and RIG-I activations were significantly reduced when mRNA contained modified nucleosides such as m5C, m6A, 5-methyluridine (m5U), Ψ-UTP, and 2-thiouridine (s2U).20, 150, 230, 234 RNA-dependent protein kinase (PKR) arbitrated the immune response, and translation inhibition (by phosphorylating the alpha subunit of translation initiation factor 2 [eIF-2α]) can be escaped using Ψ-UTP- or m5C-modified nucleosides.235 m5C is explicitly recognized by the mRNA export adaptor ALYREF, and it increases mRNA-binding affinity and associated mRNA export.236 Activation of two important components of the innate immune response against unmodified mRNA, the interferon-induced enzymes 2′-5′-oligoadenylate synthetase (OAS) and RNase L, can be limited by Ψ-UTP.237 N1-methylpseudouridine (N1-mΨ-UTP) is the most used chemical modification in recent studies, and it showed remarkable expression compared to Ψ-UTP-substituted mRNA, even when delivered by different routes in vivo.238 N1-mΨ-UTP induces a tight binding to RIG-I but failed to activate RIG-I signaling (Figure 2).234, 239

Figure 2.

Figure 2

Processing of IVT mRNA in a Cell

(A) In vitro-transcribed (IVT) mRNA from linearized DNA or PCR-amplified fragment is used to transfect the cell of interest. Step 1: mRNA protection from RNase degradation and mRNA uptake are facilitated by various carriers. Step 2 of mRNA transport and release inside the cell is still unclear. Different capping modification can increase translation in step 3 and also protect from degradation. In steps 3 and 4, the translated protein from delivered mRNA gets transferred to various parts of the cell system based on post-translational modification. For an immunotherapeutic approach, the translated protein needs to get degraded by proteasome to antigen epitopes and delivered to MHC (major histocompatibility complex) class I located in the endoplasmic reticulum. MHC class I mediates surface presentation of the presented epitope to CD8+ cytotoxic T cells.320 The T cell further initiates the immune response by relocating the antigen and presenting in to MHC II. (B) IVT mRNA cause inflammatory responses and inhibition of mRNA replication as triphosphorylated mRNA or double-stranded RNA (dsRNA) can be recognized by Toll-like receptors 3, 7, and 8 (endosomal innate immune receptors), which can initiate inflammation associated with type 1 interferon (IFN), interleukin-6 and -12, and tumor necrosis factor (TNF).230 Cytoplasmic receptors, protein kinase R (PKR), retinoic acid-inducible gene I protein (RIG-I), melanoma differentiation-associated protein (MDA5), and 2′-5′-oligoadenylate synthase (OAS) can detect triphosphorylated mRNA or dsRNA and stalled translation through eIF2α, RNA degradation by ribonuclease L (RNase L), and inhibition of mRNA replication by IFN.323, 324

Codon Optimization

Codon optimization of mRNA uses the degeneracy of the genetic code to substitute specific nucleosides of a mRNA sequence without altering the resulting amino acid composition. Several recent studies have reported a high expression by codon optimization of unmodified and Ψ-UTP-modified mRNA through enriching guanosine and/or cytosine (GC).231, 240 Cas9 activity has been reported to produce significantly higher insertion or deletion (indel) and to be less immunogenic when uridine depletion has been used with 5-methoxyuridine (5moU) modification compared to unmodified and Ψ-UTP-modified Cas9 mRNA.215 Pharmacologically favorable mRNA that has undergone modification and sequence optimization still needs to be transferred in lung using carriers, to circumvent the naturally occurring barriers the lung possesses.

Reaching the Lung

In vivo delivery of mRNA therapeutics remains one of the biggest hurdles for mRNA-based therapies in general. Apart from the fragility of the mRNA molecules and the ubiquitous existence of RNases, there are two main obstacles in the delivery of mRNA in vivo: targeting specific lung cell populations and crossing the cellular membrane. For solving the latter part, nanocarriers as a delivery system have gained increased attention. Therefore, this review focuses on the benefits and obstacles in the use of mRNA-nanocarrier complexes.

In terms of transport of mRNA to a specific tissue, the route of administration plays an important role when discussing the hurdles of organ-specific mRNA therapy. Focusing on the lung, two main routes of administration were investigated in the past: i.v. application of mRNA or mRNA-complexes to reach the lung from the vascular structures and i.t. delivery of mRNA therapeutics via dry powder insufflation or high-pressure liquid suspensions using a microsprayer.150, 166, 241, 242, 243, 244, 245, 246

Intravenous application of RNAs leads to a systemic distribution of the therapeutic throughout the whole organism.247 This can be beneficial for certain pulmonary diseases like CF affecting other organs; on the contrary, only a small percentage of an active agent reaches the desired location.239 A substantial amount of mRNA is removed from the bloodstream, especially in the liver and spleen.248 This increases the amount of mRNA needed for reaching effective dose levels in the lung.20 Another hurdle emerges in the lung itself. The lung has a capillary system that consists of mostly small and non-fenestrated capillaries.249 This is very efficient for gas exchange at the alveoli, but it does not permit a free exchange of larger molecules out of the blood into the tissue. This phenomenon affects mRNA therapeutics by reaching cells in close proximity, while cells farther away from capillaries are harder to reach by i.v. administration.250 In general, i.v. application of mRNA has the advantage of circumventing some initial innate defense systems and lung barriers while fighting with the problems of systemic application of dispersed distribution and losing the targeted administration toward the lung.

In contrast, i.t. administration of therapeutic substances gives the advantage of local application of mRNA in the lung and airways. Moreover, the alveolar surface area is large and suitable for drug absorption, and the epithelial barrier is thin,242 which could facilitate the delivery of mRNA to lung cells. While endothelial cells and lung stem cells are difficult to access, alveolar cells, epithelial cells, and macrophages can be targeted by i.t. administration.251 A potential therapeutic has to be appropriately formulated to be able to reach deep lung surfaces. This includes a particle size of 4–7 μm in diameter252 for targeting the tracheobronchial region and 1–3 μm for targeting the alveolar region, when preparing a powder for insufflation or nebulizing a liquid (Figure 3).253

Figure 3.

Figure 3

Deposition of Nanoparticles for Delivery in the Lung after Intratracheal or Intravenous Administration

Left: intratracheal instillation requires a particle size of 1–3 μm to reach the alveoli efficiently; particles from 4 to 7 μm are mainly distributed to the upper airways and main bronchioles, and particles smaller than 1 μm are exhaled again.252, 318 Right: inhaled nanoparticles can enter bronchial as well as alveolar epithelium; nanoparticles can enter lymph and blood circulation to be delivered to secondary organs.319 Intravenous injection can systemically deliver nanoparticles to a limited part of the alveolar epithelium due to small and non-fenestrated endothelial cells in the capillaries in the lung.250

To reach deep lung structures and alveoli, mRNA therapeutics still has to pass the respiratory mucus. In a non-pathological condition, the thickness of the mucus is between 2 and 5 μm in the bronchi and 10 and 30 μm in the trachea,254 while in CF asthma and COPD the mucus layer is reported to be much thicker.255 The gel-on-brush model of Button et al.256 states that ciliary movement transports all material out of the lung at a rate of 3.6 mm/min,257 while a layer of mucins and glycoproteins form a fine mesh preventing large particles from reaching the periciliary layer and epithelial cells in the lung.256 The mucus layer, lining epithelial cells from the nose to the terminal bronchioles, also affects nanocarriers by sterical obstruction or direct interaction from diffusion to the target cells.251 Different independent studies showed a correlation between nanocarrier size and mobility in respiratory mucus. Sanders et al.258 and Dawson et al.259 reported that nanospheres at the size of ∼100 nm were able to pass more or less freely through CF sputum compared to nanospheres larger than 500 nm. Broughton-Head et al.260 detected that CF sputa of three CF patients contained a mesh with a mean size of 300 ± 106 nm, 578 ± 191 nm, and 711 ± 328 nm, respectively, providing evidence for the distinct transport parameters of different-sized nanospheres. Studies by Stern et al.,261 Kitson et al., 262 and Ferrari et al.263 suggested that not only the retention by the mesh structure in the CF sputum but also the direct interaction with free DNA present in CF sputum can reduce the gene transfer of 3ß-[N-(N',N'-dimethylaminoethane)-carbamoyl] (DC)-cholesterol (Chol)/1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE)-based lipoplexes up to 20-fold.

Respiratory mucus is not the only fluid presenting a barrier for the nanocarrier delivery of mRNA. Also, the alveolar fluid is known to inhibit cationic lipid nanocarriers, presumably by disintegration of the lipoplexes by negatively charged lipids in the surfactant.251 In contrast, PEI and dendrimer polyamidoamine (PAMAM)-based gene delivery was observed to be resistant to the effects of pulmonary surfactant in vitro and in vivo.264, 265 Moreover, Exosurf (a synthetic surfactant) has been reported to increase the efficiency of PAMAM-pDNA complexes in vitro.265

To overcome the obstacles presented by both respiratory mucus and pulmonary surfactant, various approaches have been tested. Mucolytic agents, which degrade the biopolymer network built up out of DNA, actin, and mucins, are a focus of many research groups.258, 259, 260, 263 Recombinant human DNase (rhDNase) liquifies CF mucus by cleaving DNA chains in the biopolymer, and it has a direct effect on the mobility of nanocarriers of CF sputum. It increased the mobility of 270-nm nanospheres and 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP)/DOPE-based lipoplexes 2.5-fold and 1.4-fold, respectively.258, 266 N-Acetylcysteine (NAC), a clinically used mucolytic agent for asthma, COPD, and CF patients, reduces the disulfate bonds between mucins and lowers the viscosity and elasticity of respiratory mucus.267, 268 NAC-mediated mucus clearance of an ex vivo sheep trachea model increased gene transfer via p-ethyl-dimyristoylphosphadityl choline (EDMPC)-Chol lipoplexes and PEI-based polyplexes 20-fold and 10-fold, respectively. If the nasal epithelium of mice is treated with NAC 30 min before the administration of EDMPC-Chol lipoplexes, the gene expression can be increased up to 100-fold.263

An alternative strategy is to coat nanocarriers with biocompatible hydrophilic but biologically inert polymers251 to shield the nanocarriers from respiratory fluids and surfactant. GL67 (genzyme lipid 67)/DOPE lipoplexes can be coated with polyethylene glycol (PEG), and they have been reported to circumvent the adverse effects of CF mucus during gene transfection in vivo.269 Maisel et al.270 reported that 10–40 kDa PEG-coated nanocarriers can diffuse through the mucus as a mucoinert particle.

Nanotransporters to Target the Lung

The labile nature of mRNA and immunogenicity are the biggest hurdles of mRNA therapeutics. As discussed above, the immunogenicity has been overcome by chemical modifications; however, the instability of mRNA under physiological conditions requires additional action. Electroporation,271 gene gun,272 microinjection,273 and sonoporation274 have been investigated for mRNA delivery; however, these are restricted to ex vivo manipulation and ill suited for systemic delivery. Therefore, suitable mRNA carriers should exhibit the following functions: protection from RNase degradation, evasion of direct renal clearance, avoidance of nonspecific interaction, facilitation of mRNA stability, and sufficient mRNA loading and release.275, 276, 277 The physiochemical properties, such as hydrodynamic diameter, shape, size, surface charge, solubility, flexibility, stability, formulation, and body composition with regard to route of administration, decide the target binding bio-distribution as well as the clearance of the nanocarriers. As many materials used to construct nanoparticles are toxic or potentially toxic, biocompatibility and biodegradability become key factors. Since our focus is delivering mRNA therapeutics to the lung, we cover biomaterials such as lipids, polymers, and combined formulations that are developed for delivery to the lung.

Lipid-based nanoparticles (LNPs) or lipoplexes have gained popularity since the beginning of drug and nucleic acid delivery.9, 231, 240, 278, 279These have the significant advantages of easy synthesis, scalability, low batch variability, and biocompatibility.280, 281, 282 Commercially available lipoplexes, such as RNAiMAX, Stemfect, and Megafectin, have been successfully used in transfecting mRNA in vivo.279, 283, 284, 285 Cationic lipids, such as 1,2-di-O-octadecenyl-3-trimethylammonium-propane (DOTMA), DOTAP, and zwitterionic DOPE, have been used alone or in combination, as these readily form complexes with mRNA. Variants to reduce toxicity and immunogenicity associated with cationic lipids and with improved efficacy have been developed. Additionally, the ratio of components substantially affects LNP efficacy. These include ionizable lipidic systems that can reduce toxicity by possessing a neutral charge at physiological pH286 and ionizable lipid nanoparticles consisting of ionizable lipid, cholesterol (hydrophobic), helper lipid (DOPE or 1,2-distearoyl-sn-glycero-3-phosphocholine [DSPC]), and PEG lipids. DOPE enhances efficacy by promoting membrane fusion (cell and endosomal), and PEG lipids prevent reticuloendothelial clearance and reduce opsonization by serum proteins.

LNP-mediated mRNA delivery has been extensively used in protein replacement therapies, vaccines, and cancer immunotherapies.276 Earlier work by Litzinger et al.287 showed that cationic liposomes of size 2.0 μm are transiently taken up by the lung, followed by rapid distribution to the liver. Similarly, efficient pulmonary endothelial delivery of plasmid was achieved with a lipid vector consisting of DOTAP liposomes, protamine, and oligo deoxynucleotides,288 with minimum cytotoxicity and release of proinflammatory cytokines. The landmark clinical trial of CFTR gene therapy with pGM169179 and multiple other clinical trials with nasal delivery tested lipid nanoparticle cholest-5-en-3-ol(3β)-,3-[(3-aminopropyl)[4-[(3-aminopropyl)amino] butyl] carbamate] (GL67A) due to its desirable stability during aerosolization,289 gene transfer potency,290 and well-characterized safety parameters.291 The Wendel group292 has shown DOPE liposomes as potential transfection agents for AAT mRNA, resulting in prolonged protein production of AAT in vitro with improved stability of mRNA in liposomes for up to 80 days, without the loss of transfection efficacy. Alexion Pharmaceuticals has shown human AAT expression in both mouse liver and lung upon i.v. injection of mRNA-ionizable LNP complex after 24 h.21 Both approaches require further testing on knockout mouse models293 to check its efficiency as a therapy for AATD.

Lipid-enabled and unlocked nucleomonomer agent-modified RNA (LUNAR) technology of Arcturus Therapeutics employs biodegradable ionizable lipids (ATX, Arcturus Therapeutics’s proprietary lipid) that have shown no adverse events, hepatotoxicity, weight loss, or innate or adaptive immune reactions in response to treatment with repeated dosing of up to 4 months.294 Ramaswamy et al.294 observed faster translation (within 6 h) and major deposition of LUNAR-encapsulated mRNA in mouse liver with i.v. injection. Arcturus holds multiple patents on nanoparticles for RNA delivery with the potential of lung epithelial delivery via nebulization. Translate Bio holds a patent on multiple lipid nanoparticles with its collaborator at Massachusetts Institute of Technology (MIT) and Imperial College London, and it is the first one to enter clinical trials with LNPs (ClinicalTrials.gov: NCT03375047) for mRNA-based CF therapy. Valera (by Moderna) has reported efficacy of modified hemagglutinin mRNA-LNP-formulated vaccines against H7N9 and H10N8 influenza virus (presently at clinical phase I), when immunized intradermally or intramuscularly in mice, ferrets, and non-human primates293 (one must note that localization in lungs is not required for immunization).

Though not as equally advanced as LNPs, polymer-based nanoparticles have shown considerable potential in aiding therapeutics. Cationic polymers (linear or branched) can enable nucleic acid shuttling across membranes by compactly packing them into nanoplexes, and they can help in cellular uptake via endocytosis.295 PEI is the vastly studied polymer for gene or oligonucleotide delivery,296, 297 however, toxicity due to nondegradability, high molecular weight (>20 kDa), and its highly branched formulations has limited its clinical applications.298 The positive charge attributes to interaction with serum proteins (negatively charged), resulting in their aggregation and increase in size that causes toxicity, similar to that of cationic liposomes. Therefore, different groups have tried to modify PEI and achieve higher transfection with lower toxicity, which includes reducing size299 (mRNA release), reducing molecular weight,300 or using additives.301

Poly (L-lysine) (PLL), poly(2-(dimethylamino)ethyl methacrylate) (p[DMAEMA]), and PLGA are well known polymers, and diblock302 and triblock polymers303 have shown encouraging results for nucleic acid transfection. As already discussed, mucus acts as a strong barrier, and mucoadhesive particles can increase the residence time while bulking up the nanoparticle. PEGylated NPs (diblock copolymer composed of PLGA and PEG [PLGA-PEG], namely, PLGA-PEG mucous penetrating particle [MPP]) of >200-nm size are known to penetrate mucus and CF sputum.304, 305 Based on this, Schneider et al.305 and others have shown that MPP (≤300 nm in diameter) exhibits improved particle distribution and lung retention.150 Chitosan (mucoadhesive) coating20, 306 of PLGA nanoparticles has shown successful delivery of SP-B mRNA and hCFTR mRNA in mouse models, significantly improving survival307 and lung function,308 respectively. It is possible that these enable deeper lung delivery instead of mere epithelial delivery, due to which we could observe the survival of an SP-B-deficient mouse model309 when corrected with zinc-finger nuclease (ZFN) mRNA and donor template. In an attempt to develop nanoparticles for pulmonary delivery, Ethris used mRNA complexed with a polymer scaffold of poly (acrylic acid) of 20 kDa grafted with oligoalkylamines, which showed delivery in cranial parts of pig lung upon nebulization.54 A study about the exact localization of developed nanoparticles in different lung cell populations would benefit the scientific society in moving toward disease-specific targeted therapy for different lung diseases.

Further, combinations of lipids and polymers have been tested as nanocarriers. These include self-assembling nano-micelles formed by copolymer consisting of polyamino acid block and PEG with mRNA at core. Commonly used polyamino blocks include poly(Nʹ-(N-(2-aminoethyl)-2-aminoethyl) aspartamide (PAsp[DET]), which has shown protein expression in nasal neurons with mRNA coding for brain-derived neurotrophic factors (BDNFs).310 The complex also reduced apoptosis when injected with anti-apoptotic protein B cell lymphoma (Bcl-2) mRNA in a fulminant hepatitis mouse model.311 The group of Daniel G. Anderson has developed various nanoparticles to be used in therapeutics,14, 312 among which the polymer lipid combination of poly (β-amino esters) (PBAEs) and PEG has shown greater potential in delivering mRNA to lung via i.v. injection,313 aiding successful systemic delivery. Desrosiers et al.314 have developed amine-modified polyester-based nanocarriers in combination with triblock copolymers, with specific mRNA delivery to lung. Though degradable and optimized for serum stability and reduced toxicity, these need to be further tested for inflammatory reactions before clinical translation. Recent publication by Patel et al.315 on hyperbranched PBAEs has shown ease in nebulization and uniform distribution of mRNA in all 5 lobes of lung, with no measured local or systemic toxicity.

To further increase the specificity of nanocarrier-based delivery, receptor-based technologies have been tested. Arrowhead Pharmaceuticals developed an asialoglycoprotein receptor-targeting nanocarrier to specifically administer an RNAi molecule (targeted RNAi molecule [TRiM]) to reduce the accumulation of AAT protein230 for AAT-related liver diseases. If combined with pulmonary delivery of AAT mRNA, it can act as a complete therapy for diseases like AATD. A receptor-based method can also be developed for the lung epithelium, but specific markers of lung epithelial cells have to be identified to avoid cross-reactivity with other epithelial cell linings. In another approach for enhancing translation and reducing the degradation of mRNA upon entry, a delivery system has been tested that employs translation initiation factor eIF4E with cationic polyamine. A study showed that these nanoparticles induced mRNA expression in mouse lung upon systemic delivery.316 Other nanoparticles developed for lungs include gelatin nanocarriers crosslinked with genipin, monomethoxypoly(ethylene glycol)-poly(lactic-co-glycolic acid)-poly-l-lysine (mPEG-PLGA-PLL) triblock copolymers, MUC-1 aptamer-functionalized hybrid nanoparticles, drug-loaded liposomes, anionic PAMAM dendrimers, and a recently developed virus-inspired polymer for endosomal release (VIPER).215, 230, 238, 240, 250, 252, 317, 318, 319, 320 However, as discussed before, these must be extensively tested for compatibility with mRNA.

Conclusions

This paper focused on respiratory diseases and associated cell populations of the lung. To understand the disease pathology and possible countermeasures, the physiological aspects of various lung cells have to be determined. This includes the awareness of connections of epithelial cells with alveolar cells, serous cells (goblet cells), and also stem and progenitor cells, as lung diseases may originate from a single defect but can affect the whole lung. Recent developments already produced major advancements in therapies such as chemical modulators for CF, but they also produced therapy resistance or were only effective for certain variants of a disease. This makes mRNA-based protein supplementation therapy a viable alternative for diseases such as CF, SP-B deficiency, asthma, IPF, and COPD while offering a treatment independent of the underlying mutational status. Furthermore, stem cells of the lung can also be a target for mRNA-based CRISPR/Cas therapies that hold the potential for permanent cures for monogenetic lung diseases, such as CF, SP-B deficiency, and AATD.

The therapeutic potential of mRNA for protein supplementation therapy was widely unrecognized due to its instability for a long time. Over the last decades, an appreciation of mRNA as the molecule connecting the world of proteins and DNA is renewing the focus of research on mRNA.9, 20, 215 The research focuses on the properties of RNA to increase or modulate stability and evade immune recognition as well as delivery of mRNA specifically to the lung and other organs. The use of naturally occurring nucleoside modifications has diminished the recognition of mRNA by the innate immune system.230, 235 These modifications also help in improving the stability and expression of mRNA. This is further promoted by modifications like 5′-capping and the addition of a poly(A) tail as well as modifications in the 3′ and 5′ UTRs. Sequence optimization increases expression and lowers immunogenicity of mRNA therapeutics. The most optimized RNA still needs to reach the target cell to be expressed into a protein.

The problem of delivery consists of three main parts: (1) how to find a suitable route of administration; (2) as mRNA for protein supplementation therapy usually are of substantial size and negatively charged, they will not cross cellular membranes unfacilitated; and (3) the composition of mRNA and nanocarriers to maintain the optimal stability of mRNA nanocarrier complexes. Looking at protein supplementation therapies for the lungs, i.t. delivery gives the ability to apply mRNA therapeutics locally and concentrated in the lungs. To overcome defense barriers like the respiratory mucus and alveolar fluids as well as the cellular membranes, nanocarriers developed into a favorable type of vehicle for mRNA. To date, research indicates that small nanocarriers (∼100 nm) together with a polymer coating and the potential application of a mucolytic agent can improve the stability of the mRNA nanocarrier complex and mRNA uptake into the target cell. Nonetheless, the mechanism of how a nanocarrier facilitates the cellular uptake of mRNA is still not fully understood.

All in all, the pieces needed for an effective protein supplementation therapy in the lung are available in various preclinical and clinical stages. The next task is to find a formulation to bring all of these components together—route of administration, carrier, mRNA sequence and modification—to form a working therapy for patients. The first clinical trials in the slowly evolving field of protein supplementation therapy show that the concept is viable (ClinicalTrials.gov: NCT02935712 and NCT03375047; see also Table 1).

Author Contributions

I.S., A.K.M.A.H., and B.W. wrote the main parts of the article and produced graphics. P.W. helped in the meticulous preparation of the manuscript. M.S.D.K. drafted the final version of the manuscript. All authors read and approved the final manuscript.

Conflicts of Interest

M.S.D.K. holds a patent on RNA modification (EP2459231B1). M.S.D.K. and A.K.M.A.H. hold a European patent on delivery of hCFTR cmRNA complexed with nanoparticles (17169561.2-1401).

Acknowledgments

This work was supported by the European Research Council (ERC Starting Grant to M.S.D.K., 637752 “BREATHE”) and the HMZ Privatstiftung to M.S.D.K. We thank Melanie Perreault for proofreading as a native English speaker.

References

  • 1.Robertson M.P., Joyce G.F. The origins of the RNA world. Cold Spring Harb. Perspect. Biol. 2012;4:a003608. doi: 10.1101/cshperspect.a003608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Cech T.R. The RNA worlds in context. Cold Spring Harb. Perspect. Biol. 2012;4:a006742. doi: 10.1101/cshperspect.a006742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Neveu M., Kim H.J., Benner S.A. The “strong” RNA world hypothesis: fifty years old. Astrobiology. 2013;13:391–403. doi: 10.1089/ast.2012.0868. [DOI] [PubMed] [Google Scholar]
  • 4.Murgola E.J., Prather N.E., Mims B.H., Pagel F.T., Hijazi K.A. Anticodon shift in tRNA: a novel mechanism in missense and nonsense suppression. Proc. Natl. Acad. Sci. USA. 1983;80:4936–4939. doi: 10.1073/pnas.80.16.4936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Ellington A.D., Szostak J.W. In vitro selection of RNA molecules that bind specific ligands. Nature. 1990;346:818–822. doi: 10.1038/346818a0. [DOI] [PubMed] [Google Scholar]
  • 6.Cogoni C., Irelan J.T., Schumacher M., Schmidhauser T.J., Selker E.U., Macino G. Transgene silencing of the al-1 gene in vegetative cells of Neurospora is mediated by a cytoplasmic effector and does not depend on DNA-DNA interactions or DNA methylation. EMBO J. 1996;15:3153–3163. [PMC free article] [PubMed] [Google Scholar]
  • 7.Chen X., He L., Zhao Y., Li Y., Zhang S., Sun K., So K., Chen F., Zhou L., Lu L. Malat1 regulates myogenic differentiation and muscle regeneration through modulating MyoD transcriptional activity. Cell Discov. 2017;3:17002. doi: 10.1038/celldisc.2017.2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Lundstrom K. Special Issue: Gene Therapy with Emphasis on RNA Interference. Viruses. 2015;7:4482–4487. doi: 10.3390/v7082830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Malone R.W., Felgner P.L., Verma I.M. Cationic liposome-mediated RNA transfection. Proc. Natl. Acad. Sci. USA. 1989;86:6077–6081. doi: 10.1073/pnas.86.16.6077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Wolff J.A., Malone R.W., Williams P., Chong W., Acsadi G., Jani A., Felgner P.L. Direct gene transfer into mouse muscle in vivo. Science. 1990;247:1465–1468. doi: 10.1126/science.1690918. [DOI] [PubMed] [Google Scholar]
  • 11.Jirikowski G.F., Sanna P.P., Maciejewski-Lenoir D., Bloom F.E. Reversal of diabetes insipidus in Brattleboro rats: intrahypothalamic injection of vasopressin mRNA. Science. 1992;255:996–998. doi: 10.1126/science.1546298. [DOI] [PubMed] [Google Scholar]
  • 12.Newbury S.F. Control of mRNA stability in eukaryotes. Biochem. Soc. Trans. 2006;34:30–34. doi: 10.1042/BST20060030. [DOI] [PubMed] [Google Scholar]
  • 13.Mockey M., Gonçalves C., Dupuy F.P., Lemoine F.M., Pichon C., Midoux P. mRNA transfection of dendritic cells: synergistic effect of ARCA mRNA capping with Poly(A) chains in cis and in trans for a high protein expression level. Biochem. Biophys. Res. Commun. 2006;340:1062–1068. doi: 10.1016/j.bbrc.2005.12.105. [DOI] [PubMed] [Google Scholar]
  • 14.Holtkamp S., Kreiter S., Selmi A., Simon P., Koslowski M., Huber C., Türeci O., Sahin U. Modification of antigen-encoding RNA increases stability, translational efficacy, and T-cell stimulatory capacity of dendritic cells. Blood. 2006;108:4009–4017. doi: 10.1182/blood-2006-04-015024. [DOI] [PubMed] [Google Scholar]
  • 15.Elango N., Elango S., Shivshankar P., Katz M.S. Optimized transfection of mRNA transcribed from a d(A/T)100 tail-containing vector. Biochem. Biophys. Res. Commun. 2005;330:958–966. doi: 10.1016/j.bbrc.2005.03.067. [DOI] [PubMed] [Google Scholar]
  • 16.Rozenski J., Crain P.F., McCloskey J.A. The RNA Modification Database: 1999 update. Nucleic Acids Res. 1999;27:196–197. doi: 10.1093/nar/27.1.196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Boccaletto P., Machnicka M.A., Purta E., Piatkowski P., Baginski B., Wirecki T.K., de Crécy-Lagard V., Ross R., Limbach P.A., Kotter A. MODOMICS: a database of RNA modification pathways. 2017 update. Nucleic Acids Res. 2018;46:D303–D307. doi: 10.1093/nar/gkx1030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Ishii K.J., Akira S. TLR ignores methylated RNA? Immunity. 2005;23:111–113. doi: 10.1016/j.immuni.2005.08.003. [DOI] [PubMed] [Google Scholar]
  • 19.Karikó K., Ni H., Capodici J., Lamphier M., Weissman D. mRNA is an endogenous ligand for Toll-like receptor 3. J. Biol. Chem. 2004;279:12542–12550. doi: 10.1074/jbc.M310175200. [DOI] [PubMed] [Google Scholar]
  • 20.Haque A.K.M.A., Dewerth A., Antony J.S., Riethmüller J., Schweizer G.R., Weinmann P., Latifi N., Yasar H., Pedemonte N., Sondo E. Chemically modified hCFTR mRNAs recuperate lung function in a mouse model of cystic fibrosis. Sci. Rep. 2018;8:16776. doi: 10.1038/s41598-018-34960-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Connolly B., Isaacs C., Cheng L., Asrani K.H., Subramanian R.R. SERPINA1 mRNA as a Treatment for Alpha-1 Antitrypsin Deficiency. J. Nucleic Acids. 2018;2018:8247935. doi: 10.1155/2018/8247935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Plasschaert L.W., Žilionis R., Choo-Wing R., Savova V., Knehr J., Roma G., Klein A.M., Jaffe A.B. A single-cell atlas of the airway epithelium reveals the CFTR-rich pulmonary ionocyte. Nature. 2018;560:377–381. doi: 10.1038/s41586-018-0394-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Montoro D.T., Haber A.L., Biton M., Vinarsky V., Lin B., Birket S.E., Yuan F., Chen S., Leung H.M., Villoria J. A revised airway epithelial hierarchy includes CFTR-expressing ionocytes. Nature. 2018;560:319–324. doi: 10.1038/s41586-018-0393-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Castranova V., Rabovsky J., Tucker J.H., Miles P.R. The alveolar type II epithelial cell: a multifunctional pneumocyte. Toxicol. Appl. Pharmacol. 1988;93:472–483. doi: 10.1016/0041-008x(88)90051-8. [DOI] [PubMed] [Google Scholar]
  • 25.Noguchi A., Reddy R., Kursar J.D., Parks W.C., Mecham R.P. Smooth muscle isoactin and elastin in fetal bovine lung. Exp. Lung Res. 1989;15:537–552. doi: 10.3109/01902148909069617. [DOI] [PubMed] [Google Scholar]
  • 26.Morrisey E.E., Hogan B.L. Preparing for the first breath: genetic and cellular mechanisms in lung development. Dev. Cell. 2010;18:8–23. doi: 10.1016/j.devcel.2009.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Boström H., Willetts K., Pekny M., Levéen P., Lindahl P., Hedstrand H., Pekna M., Hellström M., Gebre-Medhin S., Schalling M. PDGF-A signaling is a critical event in lung alveolar myofibroblast development and alveogenesis. Cell. 1996;85:863–873. doi: 10.1016/s0092-8674(00)81270-2. [DOI] [PubMed] [Google Scholar]
  • 28.Hilliard T.N., Regamey N., Shute J.K., Nicholson A.G., Alton E.W., Bush A., Davies J.C. Airway remodelling in children with cystic fibrosis. Thorax. 2007;62:1074–1080. doi: 10.1136/thx.2006.074641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Adam D., Roux-Delrieu J., Luczka E., Bonnomet A., Lesage J., Mérol J.C., Polette M., Abély M., Coraux C. Cystic fibrosis airway epithelium remodelling: involvement of inflammation. J. Pathol. 2015;235:408–419. doi: 10.1002/path.4471. [DOI] [PubMed] [Google Scholar]
  • 30.Elias J.A., Zhu Z., Chupp G., Homer R.J. Airway remodeling in asthma. J. Clin. Invest. 1999;104:1001–1006. doi: 10.1172/JCI8124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Fehrenbach H., Wagner C., Wegmann M. Airway remodeling in asthma: what really matters. Cell Tissue Res. 2017;367:551–569. doi: 10.1007/s00441-016-2566-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Knudsen L., Ruppert C., Ochs M. Tissue remodelling in pulmonary fibrosis. Cell Tissue Res. 2017;367:607–626. doi: 10.1007/s00441-016-2543-2. [DOI] [PubMed] [Google Scholar]
  • 33.Wallace W.A., Fitch P.M., Simpson A.J., Howie S.E. Inflammation-associated remodelling and fibrosis in the lung - a process and an end point. Int. J. Exp. Pathol. 2007;88:103–110. doi: 10.1111/j.1365-2613.2006.00515.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Pain M., Bermudez O., Lacoste P., Royer P.J., Botturi K., Tissot A., Brouard S., Eickelberg O., Magnan A. Tissue remodelling in chronic bronchial diseases: from the epithelial to mesenchymal phenotype. Eur. Respir. Rev. 2014;23:118–130. doi: 10.1183/09059180.00004413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Salazar L.M., Herrera A.M. Fibrotic response of tissue remodeling in COPD. Hai. 2011;189:101–109. doi: 10.1007/s00408-011-9279-2. [DOI] [PubMed] [Google Scholar]
  • 36.Sato M., Keshavjee S. Bronchiolitis obliterans syndrome: alloimmune-dependent and -independent injury with aberrant tissue remodeling. Semin. Thorac. Cardiovasc. Surg. 2008;20:173–182. doi: 10.1053/j.semtcvs.2008.05.002. [DOI] [PubMed] [Google Scholar]
  • 37.Ma J., Rubin B.K., Voynow J.A. Mucins, Mucus, and Goblet Cells. Chest. 2018;154:169–176. doi: 10.1016/j.chest.2017.11.008. [DOI] [PubMed] [Google Scholar]
  • 38.Chen G., Korfhagen T.R., Xu Y., Kitzmiller J., Wert S.E., Maeda Y., Gregorieff A., Clevers H., Whitsett J.A. SPDEF is required for mouse pulmonary goblet cell differentiation and regulates a network of genes associated with mucus production. J. Clin. Invest. 2009;119:2914–2924. doi: 10.1172/JCI39731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Rajavelu P., Chen G., Xu Y., Kitzmiller J.A., Korfhagen T.R., Whitsett J.A. Airway epithelial SPDEF integrates goblet cell differentiation and pulmonary Th2 inflammation. J. Clin. Invest. 2015;125:2021–2031. doi: 10.1172/JCI79422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.De Rose V., Molloy K., Gohy S., Pilette C., Greene C.M. Airway Epithelium Dysfunction in Cystic Fibrosis and COPD. Mediators Inflamm. 2018;2018:1309746. doi: 10.1155/2018/1309746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Wynn T.A., Cheever A.W., Jankovic D., Poindexter R.W., Caspar P., Lewis F.A., Sher A. An IL-12-based vaccination method for preventing fibrosis induced by schistosome infection. Nature. 1995;376:594–596. doi: 10.1038/376594a0. [DOI] [PubMed] [Google Scholar]
  • 42.Whitsett J.A. Airway Epithelial Differentiation and Mucociliary Clearance. Ann. Am. Thorac. Soc. 2018;15(Suppl 3):S143–S148. doi: 10.1513/AnnalsATS.201802-128AW. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Gerbe F., Sidot E., Smyth D.J., Ohmoto M., Matsumoto I., Dardalhon V., Cesses P., Garnier L., Pouzolles M., Brulin B. Intestinal epithelial tuft cells initiate type 2 mucosal immunity to helminth parasites. Nature. 2016;529:226–230. doi: 10.1038/nature16527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Tizzano M., Gulbransen B.D., Vandenbeuch A., Clapp T.R., Herman J.P., Sibhatu H.M., Churchill M.E., Silver W.L., Kinnamon S.C., Finger T.E. Nasal chemosensory cells use bitter taste signaling to detect irritants and bacterial signals. Proc. Natl. Acad. Sci. USA. 2010;107:3210–3215. doi: 10.1073/pnas.0911934107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Krasteva G., Canning B.J., Hartmann P., Veres T.Z., Papadakis T., Mühlfeld C., Schliecker K., Tallini Y.N., Braun A., Hackstein H. Cholinergic chemosensory cells in the trachea regulate breathing. Proc. Natl. Acad. Sci. USA. 2011;108:9478–9483. doi: 10.1073/pnas.1019418108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Gordon E.D., Locksley R.M., Fahy J.V. Cross-Talk between Epithelial Cells and Type 2 Immune Signaling. The Role of IL-25. Am. J. Respir. Crit. Care Med. 2016;193:935–936. doi: 10.1164/rccm.201512-2534ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Zhang Y.J., O’Neal W.K., Randell S.H., Blackburn K., Moyer M.B., Boucher R.C., Ostrowski L.E. Identification of dynein heavy chain 7 as an inner arm component of human cilia that is synthesized but not assembled in a case of primary ciliary dyskinesia. J. Biol. Chem. 2002;277:17906–17915. doi: 10.1074/jbc.M200348200. [DOI] [PubMed] [Google Scholar]
  • 48.Ostrowski L.E., Blackburn K., Radde K.M., Moyer M.B., Schlatzer D.M., Moseley A., Boucher R.C. A proteomic analysis of human cilia: identification of novel components. Mol. Cell. Proteomics. 2002;1:451–465. doi: 10.1074/mcp.m200037-mcp200. [DOI] [PubMed] [Google Scholar]
  • 49.Ostrowski L.E., Dutcher S.K., Lo C.W. Cilia and models for studying structure and function. Proc. Am. Thorac. Soc. 2011;8:423–429. doi: 10.1513/pats.201103-027SD. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Bustamante-Marin X.M., Ostrowski L.E. Cilia and Mucociliary Clearance. Cold Spring Harb. Perspect. Biol. 2017;9:a028241. doi: 10.1101/cshperspect.a028241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Whitsett J.A., Alenghat T. Respiratory epithelial cells orchestrate pulmonary innate immunity. Nat. Immunol. 2015;16:27–35. doi: 10.1038/ni.3045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Sanderson M.J., Dirksen E.R. Mechanosensitivity of cultured ciliated cells from the mammalian respiratory tract: implications for the regulation of mucociliary transport. Proc. Natl. Acad. Sci. USA. 1986;83:7302–7306. doi: 10.1073/pnas.83.19.7302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Shah A.S., Ben-Shahar Y., Moninger T.O., Kline J.N., Welsh M.J. Motile cilia of human airway epithelia are chemosensory. Science. 2009;325:1131–1134. doi: 10.1126/science.1173869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Horani A., Ferkol T.W., Dutcher S.K., Brody S.L. Genetics and biology of primary ciliary dyskinesia. Paediatr. Respir. Rev. 2016;18:18–24. doi: 10.1016/j.prrv.2015.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Yaghi A., Zaman A., Cox G., Dolovich M.B. Ciliary beating is depressed in nasal cilia from chronic obstructive pulmonary disease subjects. Respir. Med. 2012;106:1139–1147. doi: 10.1016/j.rmed.2012.04.001. [DOI] [PubMed] [Google Scholar]
  • 56.Tamashiro E., Xiong G., Anselmo-Lima W.T., Kreindler J.L., Palmer J.N., Cohen N.A. Cigarette smoke exposure impairs respiratory epithelial ciliogenesis. Am. J. Rhinol. Allergy. 2009;23:117–122. doi: 10.2500/ajra.2009.23.3280. [DOI] [PubMed] [Google Scholar]
  • 57.Simet S.M., Sisson J.H., Pavlik J.A., Devasure J.M., Boyer C., Liu X., Kawasaki S., Sharp J.G., Rennard S.I., Wyatt T.A. Long-term cigarette smoke exposure in a mouse model of ciliated epithelial cell function. Am. J. Respir. Cell Mol. Biol. 2010;43:635–640. doi: 10.1165/rcmb.2009-0297OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Guichard C., Harricane M.C., Lafitte J.J., Godard P., Zaegel M., Tack V., Lalau G., Bouvagnet P. Axonemal dynein intermediate-chain gene (DNAI1) mutations result in situs inversus and primary ciliary dyskinesia (Kartagener syndrome) Am. J. Hum. Genet. 2001;68:1030–1035. doi: 10.1086/319511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Olbrich H., Häffner K., Kispert A., Völkel A., Volz A., Sasmaz G., Reinhardt R., Hennig S., Lehrach H., Konietzko N. Mutations in DNAH5 cause primary ciliary dyskinesia and randomization of left-right asymmetry. Nat. Genet. 2002;30:143–144. doi: 10.1038/ng817. [DOI] [PubMed] [Google Scholar]
  • 60.Pifferi M., Michelucci A., Conidi M.E., Cangiotti A.M., Simi P., Macchia P., Boner A.L. New DNAH11 mutations in primary ciliary dyskinesia with normal axonemal ultrastructure. Eur. Respir. J. 2010;35:1413–1416. doi: 10.1183/09031936.00186209. [DOI] [PubMed] [Google Scholar]
  • 61.Hjeij R., Lindstrand A., Francis R., Zariwala M.A., Liu X., Li Y., Damerla R., Dougherty G.W., Abouhamed M., Olbrich H. ARMC4 mutations cause primary ciliary dyskinesia with randomization of left/right body asymmetry. Am. J. Hum. Genet. 2013;93:357–367. doi: 10.1016/j.ajhg.2013.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Duriez B., Duquesnoy P., Escudier E., Bridoux A.M., Escalier D., Rayet I., Marcos E., Vojtek A.M., Bercher J.F., Amselem S. A common variant in combination with a nonsense mutation in a member of the thioredoxin family causes primary ciliary dyskinesia. Proc. Natl. Acad. Sci. USA. 2007;104:3336–3341. doi: 10.1073/pnas.0611405104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Horani A., Druley T.E., Zariwala M.A., Patel A.C., Levinson B.T., Van Arendonk L.G., Thornton K.C., Giacalone J.C., Albee A.J., Wilson K.S. Whole-exome capture and sequencing identifies HEATR2 mutation as a cause of primary ciliary dyskinesia. Am. J. Hum. Genet. 2012;91:685–693. doi: 10.1016/j.ajhg.2012.08.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Olbrich H., Schmidts M., Werner C., Onoufriadis A., Loges N.T., Raidt J., Banki N.F., Shoemark A., Burgoyne T., Al Turki S., UK10K Consortium Recessive HYDIN mutations cause primary ciliary dyskinesia without randomization of left-right body asymmetry. Am. J. Hum. Genet. 2012;91:672–684. doi: 10.1016/j.ajhg.2012.08.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Merveille A.C., Davis E.E., Becker-Heck A., Legendre M., Amirav I., Bataille G., Belmont J., Beydon N., Billen F., Clément A. CCDC39 is required for assembly of inner dynein arms and the dynein regulatory complex and for normal ciliary motility in humans and dogs. Nat. Genet. 2011;43:72–78. doi: 10.1038/ng.726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Becker-Heck A., Zohn I.E., Okabe N., Pollock A., Lenhart K.B., Sullivan-Brown J., McSheene J., Loges N.T., Olbrich H., Haeffner K. The coiled-coil domain containing protein CCDC40 is essential for motile cilia function and left-right axis formation. Nat. Genet. 2011;43:79–84. doi: 10.1038/ng.727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Horani A., Brody S.L., Ferkol T.W., Shoseyov D., Wasserman M.G., Ta-shma A., Wilson K.S., Bayly P.V., Amirav I., Cohen-Cymberknoh M. CCDC65 mutation causes primary ciliary dyskinesia with normal ultrastructure and hyperkinetic cilia. PLoS ONE. 2013;8:e72299. doi: 10.1371/journal.pone.0072299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Panizzi J.R., Becker-Heck A., Castleman V.H., Al-Mutairi D.A., Liu Y., Loges N.T., Pathak N., Austin-Tse C., Sheridan E., Schmidts M. CCDC103 mutations cause primary ciliary dyskinesia by disrupting assembly of ciliary dynein arms. Nat. Genet. 2012;44:714–719. doi: 10.1038/ng.2277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Knowles M.R., Leigh M.W., Ostrowski L.E., Huang L., Carson J.L., Hazucha M.J., Yin W., Berg J.S., Davis S.D., Dell S.D., Genetic Disorders of Mucociliary Clearance Consortium Exome sequencing identifies mutations in CCDC114 as a cause of primary ciliary dyskinesia. Am. J. Hum. Genet. 2013;92:99–106. doi: 10.1016/j.ajhg.2012.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Hjeij R., Onoufriadis A., Watson C.M., Slagle C.E., Klena N.T., Dougherty G.W., Kurkowiak M., Loges N.T., Diggle C.P., Morante N.F., UK10K Consortium CCDC151 mutations cause primary ciliary dyskinesia by disruption of the outer dynein arm docking complex formation. Am. J. Hum. Genet. 2014;95:257–274. doi: 10.1016/j.ajhg.2014.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Loges N.T., Olbrich H., Becker-Heck A., Häffner K., Heer A., Reinhard C., Schmidts M., Kispert A., Zariwala M.A., Leigh M.W. Deletions and point mutations of LRRC50 cause primary ciliary dyskinesia due to dynein arm defects. Am. J. Hum. Genet. 2009;85:883–889. doi: 10.1016/j.ajhg.2009.10.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Omran H., Kobayashi D., Olbrich H., Tsukahara T., Loges N.T., Hagiwara H., Zhang Q., Leblond G., O’Toole E., Hara C. Ktu/PF13 is required for cytoplasmic pre-assembly of axonemal dyneins. Nature. 2008;456:611–616. doi: 10.1038/nature07471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Mitchison H.M., Schmidts M., Loges N.T., Freshour J., Dritsoula A., Hirst R.A., O’Callaghan C., Blau H., Al Dabbagh M., Olbrich H. Mutations in axonemal dynein assembly factor DNAAF3 cause primary ciliary dyskinesia. Nat. Genet. 2012;44:381–389. doi: 10.1038/ng.1106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Castleman V.H., Romio L., Chodhari R., Hirst R.A., de Castro S.C., Parker K.A., Ybot-Gonzalez P., Emes R.D., Wilson S.W., Wallis C. Mutations in radial spoke head protein genes RSPH9 and RSPH4A cause primary ciliary dyskinesia with central-microtubular-pair abnormalities. Am. J. Hum. Genet. 2009;84:197–209. doi: 10.1016/j.ajhg.2009.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Tarkar A., Loges N.T., Slagle C.E., Francis R., Dougherty G.W., Tamayo J.V., Shook B., Cantino M., Schwartz D., Jahnke C., UK10K DYX1C1 is required for axonemal dynein assembly and ciliary motility. Nat. Genet. 2013;45:995–1003. doi: 10.1038/ng.2707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Horani A., Ferkol T.W., Shoseyov D., Wasserman M.G., Oren Y.S., Kerem B., Amirav I., Cohen-Cymberknoh M., Dutcher S.K., Brody S.L. LRRC6 mutation causes primary ciliary dyskinesia with dynein arm defects. PLoS ONE. 2013;8:e59436. doi: 10.1371/journal.pone.0059436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Zariwala M.A., Gee H.Y., Kurkowiak M., Al-Mutairi D.A., Leigh M.W., Hurd T.W., Hjeij R., Dell S.D., Chaki M., Dougherty G.W. ZMYND10 is mutated in primary ciliary dyskinesia and interacts with LRRC6. Am. J. Hum. Genet. 2013;93:336–345. doi: 10.1016/j.ajhg.2013.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Wallmeier J., Al-Mutairi D.A., Chen C.T., Loges N.T., Pennekamp P., Menchen T., Ma L., Shamseldin H.E., Olbrich H., Dougherty G.W. Mutations in CCNO result in congenital mucociliary clearance disorder with reduced generation of multiple motile cilia. Nat. Genet. 2014;46:646–651. doi: 10.1038/ng.2961. [DOI] [PubMed] [Google Scholar]
  • 79.Boon M., Wallmeier J., Ma L., Loges N.T., Jaspers M., Olbrich H., Dougherty G.W., Raidt J., Werner C., Amirav I. MCIDAS mutations result in a mucociliary clearance disorder with reduced generation of multiple motile cilia. Nat. Commun. 2014;5:4418. doi: 10.1038/ncomms5418. [DOI] [PubMed] [Google Scholar]
  • 80.Rokicki W., Rokicki M., Wojtacha J., Dżeljijli A. The role and importance of club cells (Clara cells) in the pathogenesis of some respiratory diseases. Kardiochir. Torakochirurgia Pol. 2016;13:26–30. doi: 10.5114/kitp.2016.58961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Zheng D., Limmon G.V., Yin L., Leung N.H., Yu H., Chow V.T., Chen J. Regeneration of alveolar type I and II cells from Scgb1a1-expressing cells following severe pulmonary damage induced by bleomycin and influenza. PLoS ONE. 2012;7:e48451. doi: 10.1371/journal.pone.0048451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Zheng D., Soh B.S., Yin L., Hu G., Chen Q., Choi H., Han J., Chow V.T., Chen J. Differentiation of Club Cells to Alveolar Epithelial Cells In Vitro. Sci. Rep. 2017;7:41661. doi: 10.1038/srep41661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Guseh J.S., Bores S.A., Stanger B.Z., Zhou Q., Anderson W.J., Melton D.A., Rajagopal J. Notch signaling promotes airway mucous metaplasia and inhibits alveolar development. Development. 2009;136:1751–1759. doi: 10.1242/dev.029249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Roy M.G., Rahmani M., Hernandez J.R., Alexander S.N., Ehre C., Ho S.B., Evans C.M. Mucin production during prenatal and postnatal murine lung development. Am. J. Respir. Cell Mol. Biol. 2011;44:755–760. doi: 10.1165/rcmb.2010-0020OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Watson J.K., Rulands S., Wilkinson A.C., Wuidart A., Ousset M., Van Keymeulen A., Göttgens B., Blanpain C., Simons B.D., Rawlins E.L. Clonal Dynamics Reveal Two Distinct Populations of Basal Cells in Slow-Turnover Airway Epithelium. Cell Rep. 2015;12:90–101. doi: 10.1016/j.celrep.2015.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Boers J.E., Ambergen A.W., Thunnissen F.B. Number and proliferation of clara cells in normal human airway epithelium. Am. J. Respir. Crit. Care Med. 1999;159:1585–1591. doi: 10.1164/ajrccm.159.5.9806044. [DOI] [PubMed] [Google Scholar]
  • 87.Melton K.R., Nesslein L.L., Ikegami M., Tichelaar J.W., Clark J.C., Whitsett J.A., Weaver T.E. SP-B deficiency causes respiratory failure in adult mice. Am. J. Physiol. Lung Cell. Mol. Physiol. 2003;285:L543–L549. doi: 10.1152/ajplung.00011.2003. [DOI] [PubMed] [Google Scholar]
  • 88.Boers J.E., den Brok J.L., Koudstaal J., Arends J.W., Thunnissen F.B. Number and proliferation of neuroendocrine cells in normal human airway epithelium. Am. J. Respir. Crit. Care Med. 1996;154:758–763. doi: 10.1164/ajrccm.154.3.8810616. [DOI] [PubMed] [Google Scholar]
  • 89.Noguchi M., Sumiyama K., Morimoto M. Directed Migration of Pulmonary Neuroendocrine Cells toward Airway Branches Organizes the Stereotypic Location of Neuroepithelial Bodies. Cell Rep. 2015;13:2679–2686. doi: 10.1016/j.celrep.2015.11.058. [DOI] [PubMed] [Google Scholar]
  • 90.Youngson C., Nurse C., Yeger H., Cutz E. Oxygen sensing in airway chemoreceptors. Nature. 1993;365:153–155. doi: 10.1038/365153a0. [DOI] [PubMed] [Google Scholar]
  • 91.Livermore S., Zhou Y., Pan J., Yeger H., Nurse C.A., Cutz E. Pulmonary neuroepithelial bodies are polymodal airway sensors: evidence for CO2/H+ sensing. Am. J. Physiol. Lung Cell. Mol. Physiol. 2015;308:L807–L815. doi: 10.1152/ajplung.00208.2014. [DOI] [PubMed] [Google Scholar]
  • 92.Branchfield K., Nantie L., Verheyden J.M., Sui P., Wienhold M.D., Sun X. Pulmonary neuroendocrine cells function as airway sensors to control lung immune response. Science. 2016;351:707–710. doi: 10.1126/science.aad7969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Nevel R.J., Garnett E.T., Worrell J.A., Morton R.L., Nogee L.M., Blackwell T.S., Young L.R. Persistent Lung Disease in Adults with NKX2.1 Mutation and Familial Neuroendocrine Cell Hyperplasia of Infancy. Ann. Am. Thorac. Soc. 2016;13:1299–1304. doi: 10.1513/AnnalsATS.201603-155BC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Quigley I.K., Stubbs J.L., Kintner C. Specification of ion transport cells in the Xenopus larval skin. Development. 2011;138:705–714. doi: 10.1242/dev.055699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Esaki M., Hoshijima K., Nakamura N., Munakata K., Tanaka M., Ookata K., Asakawa K., Kawakami K., Wang W., Weinberg E.S., Hirose S. Mechanism of development of ionocytes rich in vacuolar-type H(+)-ATPase in the skin of zebrafish larvae. Dev. Biol. 2009;329:116–129. doi: 10.1016/j.ydbio.2009.02.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Schoch K.G., Lori A., Burns K.A., Eldred T., Olsen J.C., Randell S.H. A subset of mouse tracheal epithelial basal cells generates large colonies in vitro. Am. J. Physiol. Lung Cell. Mol. Physiol. 2004;286:L631–L642. doi: 10.1152/ajplung.00112.2003. [DOI] [PubMed] [Google Scholar]
  • 97.Borthwick D.W., Shahbazian M., Krantz Q.T., Dorin J.R., Randell S.H. Evidence for stem-cell niches in the tracheal epithelium. Am. J. Respir. Cell Mol. Biol. 2001;24:662–670. doi: 10.1165/ajrcmb.24.6.4217. [DOI] [PubMed] [Google Scholar]
  • 98.Randell S.H., Comment C.E., Ramaekers F.C., Nettesheim P. Properties of rat tracheal epithelial cells separated based on expression of cell surface alpha-galactosyl end groups. Am. J. Respir. Cell Mol. Biol. 1991;4:544–554. doi: 10.1165/ajrcmb/4.6.544. [DOI] [PubMed] [Google Scholar]
  • 99.Siebel C., Lendahl U. Notch Signaling in Development, Tissue Homeostasis, and Disease. Physiol. Rev. 2017;97:1235–1294. doi: 10.1152/physrev.00005.2017. [DOI] [PubMed] [Google Scholar]
  • 100.Morimoto M., Nishinakamura R., Saga Y., Kopan R. Different assemblies of Notch receptors coordinate the distribution of the major bronchial Clara, ciliated and neuroendocrine cells. Development. 2012;139:4365–4373. doi: 10.1242/dev.083840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Morimoto M., Liu Z., Cheng H.T., Winters N., Bader D., Kopan R. Canonical Notch signaling in the developing lung is required for determination of arterial smooth muscle cells and selection of Clara versus ciliated cell fate. J. Cell Sci. 2010;123:213–224. doi: 10.1242/jcs.058669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Hong K.U., Reynolds S.D., Watkins S., Fuchs E., Stripp B.R. In vivo differentiation potential of tracheal basal cells: evidence for multipotent and unipotent subpopulations. Am. J. Physiol. Lung Cell. Mol. Physiol. 2004;286:L643–L649. doi: 10.1152/ajplung.00155.2003. [DOI] [PubMed] [Google Scholar]
  • 103.Rock J.R., Randell S.H., Hogan B.L. Airway basal stem cells: a perspective on their roles in epithelial homeostasis and remodeling. Dis. Model. Mech. 2010;3:545–556. doi: 10.1242/dmm.006031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Araya J., Cambier S., Markovics J.A., Wolters P., Jablons D., Hill A., Finkbeiner W., Jones K., Broaddus V.C., Sheppard D. Squamous metaplasia amplifies pathologic epithelial-mesenchymal interactions in COPD patients. J. Clin. Invest. 2007;117:3551–3562. doi: 10.1172/JCI32526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Williams M.C. Alveolar type I cells: molecular phenotype and development. Annu. Rev. Physiol. 2003;65:669–695. doi: 10.1146/annurev.physiol.65.092101.142446. [DOI] [PubMed] [Google Scholar]
  • 106.Féréol S., Fodil R., Pelle G., Louis B., Isabey D. Cell mechanics of alveolar epithelial cells (AECs) and macrophages (AMs) Respir. Physiol. Neurobiol. 2008;163:3–16. doi: 10.1016/j.resp.2008.04.018. [DOI] [PubMed] [Google Scholar]
  • 107.Hsu H.S., Liu C.C., Lin J.H., Hsu T.W., Su K., Hung S.C. Repair of naphthalene-induced acute tracheal injury by basal cells depends on β-catenin. J. Thorac. Cardiovasc. Surg. 2014;148:322–332. doi: 10.1016/j.jtcvs.2013.10.039. [DOI] [PubMed] [Google Scholar]
  • 108.Hussein O., Walters B., Stroetz R., Valencia P., McCall D., Hubmayr R.D. Biophysical determinants of alveolar epithelial plasma membrane wounding associated with mechanical ventilation. Am. J. Physiol. Lung Cell. Mol. Physiol. 2013;305:L478–L484. doi: 10.1152/ajplung.00437.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Moldoveanu B., Otmishi P., Jani P., Walker J., Sarmiento X., Guardiola J., Saad M., Yu J. Inflammatory mechanisms in the lung. J. Inflamm. Res. 2009;2:1–11. [PMC free article] [PubMed] [Google Scholar]
  • 110.Krick S., Eul B.G., Hänze J., Savai R., Grimminger F., Seeger W., Rose F. Role of hypoxia-inducible factor-1alpha in hypoxia-induced apoptosis of primary alveolar epithelial type II cells. Am. J. Respir. Cell Mol. Biol. 2005;32:395–403. doi: 10.1165/rcmb.2004-0314OC. [DOI] [PubMed] [Google Scholar]
  • 111.Clark J.C., Wert S.E., Bachurski C.J., Stahlman M.T., Stripp B.R., Weaver T.E., Whitsett J.A. Targeted disruption of the surfactant protein B gene disrupts surfactant homeostasis, causing respiratory failure in newborn mice. Proc. Natl. Acad. Sci. USA. 1995;92:7794–7798. doi: 10.1073/pnas.92.17.7794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Glasser S.W., Burhans M.S., Korfhagen T.R., Na C.L., Sly P.D., Ross G.F., Ikegami M., Whitsett J.A. Altered stability of pulmonary surfactant in SP-C-deficient mice. Proc. Natl. Acad. Sci. USA. 2001;98:6366–6371. doi: 10.1073/pnas.101500298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Korfhagen T.R., Bruno M.D., Ross G.F., Huelsman K.M., Ikegami M., Jobe A.H., Wert S.E., Stripp B.R., Morris R.E., Glasser S.W. Altered surfactant function and structure in SP-A gene targeted mice. Proc. Natl. Acad. Sci. USA. 1996;93:9594–9599. doi: 10.1073/pnas.93.18.9594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Botas C., Poulain F., Akiyama J., Brown C., Allen L., Goerke J., Clements J., Carlson E., Gillespie A.M., Epstein C., Hawgood S. Altered surfactant homeostasis and alveolar type II cell morphology in mice lacking surfactant protein D. Proc. Natl. Acad. Sci. USA. 1998;95:11869–11874. doi: 10.1073/pnas.95.20.11869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Korfhagen T.R., Sheftelyevich V., Burhans M.S., Bruno M.D., Ross G.F., Wert S.E., Stahlman M.T., Jobe A.H., Ikegami M., Whitsett J.A., Fisher J.H. Surfactant protein-D regulates surfactant phospholipid homeostasis in vivo. J. Biol. Chem. 1998;273:28438–28443. doi: 10.1074/jbc.273.43.28438. [DOI] [PubMed] [Google Scholar]
  • 116.Greene K.E., Wright J.R., Steinberg K.P., Ruzinski J.T., Caldwell E., Wong W.B., Hull W., Whitsett J.A., Akino T., Kuroki Y. Serial changes in surfactant-associated proteins in lung and serum before and after onset of ARDS. Am. J. Respir. Crit. Care Med. 1999;160:1843–1850. doi: 10.1164/ajrccm.160.6.9901117. [DOI] [PubMed] [Google Scholar]
  • 117.Gregory T.J., Longmore W.J., Moxley M.A., Whitsett J.A., Reed C.R., Fowler A.A., 3rd, Hudson L.D., Maunder R.J., Crim C., Hyers T.M. Surfactant chemical composition and biophysical activity in acute respiratory distress syndrome. J. Clin. Invest. 1991;88:1976–1981. doi: 10.1172/JCI115523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Chung M.-I., Bujnis M., Barkauskas C.E., Kobayashi Y., Hogan B.L.M. Niche-mediated BMP/SMAD signaling regulates lung alveolar stem cell proliferation and differentiation. Development. 2018;145:dev163014. doi: 10.1242/dev.163014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.El-Badrawy M.K., Shalabi N.M., Mohamed M.A., Ragab A., Abdelwahab H.W. Stem Cells and Lung Regeneration. Int. J. Stem Cells. 2016;9:31–35. doi: 10.15283/ijsc.2016.9.1.31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Yei S., Bachurski C.J., Weaver T.E., Wert S.E., Trapnell B.C., Whitsett J.A. Adenoviral-mediated gene transfer of human surfactant protein B to respiratory epithelial cells. Am. J. Respir. Cell Mol. Biol. 1994;11:329–336. doi: 10.1165/ajrcmb.11.3.8086169. [DOI] [PubMed] [Google Scholar]
  • 121.Oh E.J., Mazzone S.B., Canning B.J., Weinreich D. Reflex regulation of airway sympathetic nerves in guinea-pigs. J. Physiol. 2006;573:549–564. doi: 10.1113/jphysiol.2005.104661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Lynch T.J., Anderson P.J., Rotti P.G., Tyler S.R., Crooke A.K., Choi S.H., Montoro D.T., Silverman C.L., Shahin W., Zhao R. Submucosal Gland Myoepithelial Cells Are Reserve Stem Cells That Can Regenerate Mouse Tracheal Epithelium. Cell Stem Cell. 2018;22:653–667.e5. doi: 10.1016/j.stem.2018.03.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Tata A., Kobayashi Y., Chow R.D., Tran J., Desai A., Massri A.J., McCord T.J., Gunn M.D., Tata P.R. Myoepithelial Cells of Submucosal Glands Can Function as Reserve Stem Cells to Regenerate Airways after Injury. Cell Stem Cell. 2018;22:668–683.e6. doi: 10.1016/j.stem.2018.03.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Kim C.F., Jackson E.L., Woolfenden A.E., Lawrence S., Babar I., Vogel S., Crowley D., Bronson R.T., Jacks T. Identification of bronchioalveolar stem cells in normal lung and lung cancer. Cell. 2005;121:823–835. doi: 10.1016/j.cell.2005.03.032. [DOI] [PubMed] [Google Scholar]
  • 125.Dovey J.S., Zacharek S.J., Kim C.F., Lees J.A. Bmi1 is critical for lung tumorigenesis and bronchioalveolar stem cell expansion. Proc. Natl. Acad. Sci. USA. 2008;105:11857–11862. doi: 10.1073/pnas.0803574105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Zacharek S.J., Fillmore C.M., Lau A.N., Gludish D.W., Chou A., Ho J.W., Zamponi R., Gazit R., Bock C., Jäger N. Lung stem cell self-renewal relies on BMI1-dependent control of expression at imprinted loci. Cell Stem Cell. 2011;9:272–281. doi: 10.1016/j.stem.2011.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Tropea K.A., Leder E., Aslam M., Lau A.N., Raiser D.M., Lee J.H., Balasubramaniam V., Fredenburgh L.E., Alex Mitsialis S., Kourembanas S., Kim C.F. Bronchioalveolar stem cells increase after mesenchymal stromal cell treatment in a mouse model of bronchopulmonary dysplasia. Am. J. Physiol. Lung Cell. Mol. Physiol. 2012;302:L829–L837. doi: 10.1152/ajplung.00347.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Rock J.R., Barkauskas C.E., Cronce M.J., Xue Y., Harris J.R., Liang J., Noble P.W., Hogan B.L. Multiple stromal populations contribute to pulmonary fibrosis without evidence for epithelial to mesenchymal transition. Proc. Natl. Acad. Sci. USA. 2011;108:E1475–E1483. doi: 10.1073/pnas.1117988108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Barkauskas C.E., Cronce M.J., Rackley C.R., Bowie E.J., Keene D.R., Stripp B.R., Randell S.H., Noble P.W., Hogan B.L. Type 2 alveolar cells are stem cells in adult lung. J. Clin. Invest. 2013;123:3025–3036. doi: 10.1172/JCI68782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Rawlins E.L., Okubo T., Xue Y., Brass D.M., Auten R.L., Hasegawa H., Wang F., Hogan B.L. The role of Scgb1a1+ Clara cells in the long-term maintenance and repair of lung airway, but not alveolar, epithelium. Cell Stem Cell. 2009;4:525–534. doi: 10.1016/j.stem.2009.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Whitsett J.A., Weaver T.E. Hydrophobic surfactant proteins in lung function and disease. N. Engl. J. Med. 2002;347:2141–2148. doi: 10.1056/NEJMra022387. [DOI] [PubMed] [Google Scholar]
  • 132.Nogee L.M. Alterations in SP-B and SP-C expression in neonatal lung disease. Annu. Rev. Physiol. 2004;66:601–623. doi: 10.1146/annurev.physiol.66.032102.134711. [DOI] [PubMed] [Google Scholar]
  • 133.Hartl D., Griese M. Interstitial lung disease in children -- genetic background and associated phenotypes. Respir. Res. 2005;6:32. doi: 10.1186/1465-9921-6-32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Hamvas A. Inherited surfactant protein-B deficiency and surfactant protein-C associated disease: clinical features and evaluation. Semin. Perinatol. 2006;30:316–326. doi: 10.1053/j.semperi.2005.11.002. [DOI] [PubMed] [Google Scholar]
  • 135.Hallman M., Haataja R. Genetic basis of respiratory distress syndrome. Front. Biosci. 2007;12:2670–2682. doi: 10.2741/2263. [DOI] [PubMed] [Google Scholar]
  • 136.Veldhuizen R., Nag K., Orgeig S., Possmayer F. The role of lipids in pulmonary surfactant. Biochim. Biophys. Acta. 1998;1408:90–108. doi: 10.1016/s0925-4439(98)00061-1. [DOI] [PubMed] [Google Scholar]
  • 137.Vorbroker D.K., Profitt S.A., Nogee L.M., Whitsett J.A. Aberrant processing of surfactant protein C in hereditary SP-B deficiency. Am. J. Physiol. 1995;268:L647–L656. doi: 10.1152/ajplung.1995.268.4.L647. [DOI] [PubMed] [Google Scholar]
  • 138.Li J., Ikegami M., Na C.L., Hamvas A., Espinassous Q., Chaby R., Nogee L.M., Weaver T.E., Johansson J. N-terminally extended surfactant protein (SP) C isolated from SP-B-deficient children has reduced surface activity and inhibited lipopolysaccharide binding. Biochemistry. 2004;43:3891–3898. doi: 10.1021/bi036218q. [DOI] [PubMed] [Google Scholar]
  • 139.Polin R.A., Carlo W.A., Committee on Fetus and Newborn. American Academy of Pediatrics Surfactant replacement therapy for preterm and term neonates with respiratory distress. Pediatrics. 2014;133:156–163. doi: 10.1542/peds.2013-3443. [DOI] [PubMed] [Google Scholar]
  • 140.Jobe A., Ikegami M. Surfactant for the treatment of respiratory distress syndrome. Am. Rev. Respir. Dis. 1987;136:1256–1275. doi: 10.1164/ajrccm/136.5.1256. [DOI] [PubMed] [Google Scholar]
  • 141.Hamvas A., Cole F.S., deMello D.E., Moxley M., Whitsett J.A., Colten H.R., Nogee L.M. Surfactant protein B deficiency: antenatal diagnosis and prospective treatment with surfactant replacement. J. Pediatr. 1994;125:356–361. doi: 10.1016/s0022-3476(05)83276-7. [DOI] [PubMed] [Google Scholar]
  • 142.Kendig J.W., Notter R.H., Cox C., Aschner J.L., Benn S., Bernstein R.M., Hendricks-Munoz K., Maniscalco W.M., Metlay L.A., Phelps D.L. Surfactant replacement therapy at birth: final analysis of a clinical trial and comparisons with similar trials. Pediatrics. 1988;82:756–762. [PubMed] [Google Scholar]
  • 143.Soll R.F., Hoekstra R.E., Fangman J.J., Corbet A.J., Adams J.M., James L.S., Schulze K., Oh W., Roberts J.D., Jr., Dorst J.P., Ross Collaborative Surfactant Prevention Study Group Multicenter trial of single-dose modified bovine surfactant extract (Survanta) for prevention of respiratory distress syndrome. Pediatrics. 1990;85:1092–1102. [PubMed] [Google Scholar]
  • 144.Walther F.J., Hernández-Juviel J.M., Gordon L.M., Waring A.J. Synthetic surfactant containing SP-B and SP-C mimics is superior to single-peptide formulations in rabbits with chemical acute lung injury. PeerJ. 2014;2:e393. doi: 10.7717/peerj.393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Pfister R.H., Soll R.F. New synthetic surfactants: the next generation? Biol. Neonate. 2005;87:338–344. doi: 10.1159/000084882. [DOI] [PubMed] [Google Scholar]
  • 146.van Eijk M., De Haas C.G., Haagsman H.P. Quantitative analysis of pulmonary surfactant proteins B and C. Anal. Biochem. 1995;232:231–237. doi: 10.1006/abio.1995.0012. [DOI] [PubMed] [Google Scholar]
  • 147.Seeger W., Grube C., Günther A., Schmidt R. Surfactant inhibition by plasma proteins: differential sensitivity of various surfactant preparations. Eur. Respir. J. 1993;6:971–977. [PubMed] [Google Scholar]
  • 148.Lukovic D., Plasencia I., Taberner F.J., Salgado J., Calvete J.J., Pérez-Gil J., Mingarro I. Production and characterisation of recombinant forms of human pulmonary surfactant protein C (SP-C): Structure and surface activity. Biochim. Biophys. Acta. 2006;1758:509–518. doi: 10.1016/j.bbamem.2006.03.005. [DOI] [PubMed] [Google Scholar]
  • 149.Barnett R.C., Lin X., Barravecchia M., Norman R.A., de Mesy Bentley K.L., Fazal F., Young J.L., Dean D.A. Featured Article: Electroporation-mediated gene delivery of surfactant protein B (SP-B) restores expression and improves survival in mouse model of SP-B deficiency. Exp. Biol. Med. (Maywood) 2017;242:1345–1354. doi: 10.1177/1535370217713000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Kormann M.S., Hasenpusch G., Aneja M.K., Nica G., Flemmer A.W., Herber-Jonat S., Huppmann M., Mays L.E., Illenyi M., Schams A. Expression of therapeutic proteins after delivery of chemically modified mRNA in mice. Nat. Biotechnol. 2011;29:154–157. doi: 10.1038/nbt.1733. [DOI] [PubMed] [Google Scholar]
  • 151.AstraZeneca. (2017). AstraZeneca and MedImmune enter strategic collaboration with Ethris to develop mRNA therapies for respiratory diseases. https://www.astrazeneca.com/media-centre/articles/2017/astrazeneca-and-medimmune-enter-strategic-collaboration-with-ethris-to-develop-mrna-therapies-for-respirator-diseases-17082017.html.
  • 152.Robinson D.S., Hamid Q., Ying S., Tsicopoulos A., Barkans J., Bentley A.M., Corrigan C., Durham S.R., Kay A.B. Predominant TH2-like bronchoalveolar T-lymphocyte population in atopic asthma. N. Engl. J. Med. 1992;326:298–304. doi: 10.1056/NEJM199201303260504. [DOI] [PubMed] [Google Scholar]
  • 153.Rubin B.K., Priftis K.N., Schmidt H.J., Henke M.O. Secretory hyperresponsiveness and pulmonary mucus hypersecretion. Chest. 2014;146:496–507. doi: 10.1378/chest.13-2609. [DOI] [PubMed] [Google Scholar]
  • 154.Locksley R.M. Asthma and allergic inflammation. Cell. 2010;140:777–783. doi: 10.1016/j.cell.2010.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Hellings P.W., Kasran A., Liu Z., Vandekerckhove P., Wuyts A., Overbergh L., Mathieu C., Ceuppens J.L. Interleukin-17 orchestrates the granulocyte influx into airways after allergen inhalation in a mouse model of allergic asthma. Am. J. Respir. Cell Mol. Biol. 2003;28:42–50. doi: 10.1165/rcmb.4832. [DOI] [PubMed] [Google Scholar]
  • 156.Wilson R.H., Whitehead G.S., Nakano H., Free M.E., Kolls J.K., Cook D.N. Allergic sensitization through the airway primes Th17-dependent neutrophilia and airway hyperresponsiveness. Am. J. Respir. Crit. Care Med. 2009;180:720–730. doi: 10.1164/rccm.200904-0573OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Barnes P.J. Immunology of asthma and chronic obstructive pulmonary disease. Nat. Rev. Immunol. 2008;8:183–192. doi: 10.1038/nri2254. [DOI] [PubMed] [Google Scholar]
  • 158.Finkelman F.D., Hogan S.P., Hershey G.K., Rothenberg M.E., Wills-Karp M. Importance of cytokines in murine allergic airway disease and human asthma. J. Immunol. 2010;184:1663–1674. doi: 10.4049/jimmunol.0902185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Hartl D., Koller B., Mehlhorn A.T., Reinhardt D., Nicolai T., Schendel D.J., Griese M., Krauss-Etschmann S. Quantitative and functional impairment of pulmonary CD4+CD25hi regulatory T cells in pediatric asthma. J. Allergy Clin. Immunol. 2007;119:1258–1266. doi: 10.1016/j.jaci.2007.02.023. [DOI] [PubMed] [Google Scholar]
  • 160.Karagiannidis C., Akdis M., Holopainen P., Woolley N.J., Hense G., Rückert B., Mantel P.Y., Menz G., Akdis C.A., Blaser K., Schmidt-Weber C.B. Glucocorticoids upregulate FOXP3 expression and regulatory T cells in asthma. J. Allergy Clin. Immunol. 2004;114:1425–1433. doi: 10.1016/j.jaci.2004.07.014. [DOI] [PubMed] [Google Scholar]
  • 161.Zuany-Amorim C., Sawicka E., Manlius C., Le Moine A., Brunet L.R., Kemeny D.M., Bowen G., Rook G., Walker C. Suppression of airway eosinophilia by killed Mycobacterium vaccae-induced allergen-specific regulatory T-cells. Nat. Med. 2002;8:625–629. doi: 10.1038/nm0602-625. [DOI] [PubMed] [Google Scholar]
  • 162.Arnold I.C., Dehzad N., Reuter S., Martin H., Becher B., Taube C., Müller A. Helicobacter pylori infection prevents allergic asthma in mouse models through the induction of regulatory T cells. J. Clin. Invest. 2011;121:3088–3093. doi: 10.1172/JCI45041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Doganci A., Eigenbrod T., Krug N., De Sanctis G.T., Hausding M., Erpenbeck V.J., Haddad B., Lehr H.A., Schmitt E., Bopp T. The IL-6R alpha chain controls lung CD4+CD25+ Treg development and function during allergic airway inflammation in vivo. J. Clin. Invest. 2005;115:313–325. doi: 10.1172/JCI22433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Hammad H., Kool M., Soullié T., Narumiya S., Trottein F., Hoogsteden H.C., Lambrecht B.N. Activation of the D prostanoid 1 receptor suppresses asthma by modulation of lung dendritic cell function and induction of regulatory T cells. J. Exp. Med. 2007;204:357–367. doi: 10.1084/jem.20061196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Schreiber T.H., Wolf D., Tsai M.S., Chirinos J., Deyev V.V., Gonzalez L., Malek T.R., Levy R.B., Podack E.R. Therapeutic Treg expansion in mice by TNFRSF25 prevents allergic lung inflammation. J. Clin. Invest. 2010;120:3629–3640. doi: 10.1172/JCI42933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Mays L.E., Ammon-Treiber S., Mothes B., Alkhaled M., Rottenberger J., Müller-Hermelink E.S., Grimm M., Mezger M., Beer-Hammer S., von Stebut E. Modified Foxp3 mRNA protects against asthma through an IL-10-dependent mechanism. J. Clin. Invest. 2013;123:1216–1228. doi: 10.1172/JCI65351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Curiel T.J., Coukos G., Zou L., Alvarez X., Cheng P., Mottram P., Evdemon-Hogan M., Conejo-Garcia J.R., Zhang L., Burow M. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat. Med. 2004;10:942–949. doi: 10.1038/nm1093. [DOI] [PubMed] [Google Scholar]
  • 168.Belkaid Y., Piccirillo C.A., Mendez S., Shevach E.M., Sacks D.L. CD4+CD25+ regulatory T cells control Leishmania major persistence and immunity. Nature. 2002;420:502–507. doi: 10.1038/nature01152. [DOI] [PubMed] [Google Scholar]
  • 169.Kormann M.S., Depner M., Hartl D., Klopp N., Illig T., Adamski J., Vogelberg C., Weiland S.K., von Mutius E., Kabesch M. Toll-like receptor heterodimer variants protect from childhood asthma. J. Allergy Clin. Immunol. 2008;122:86–92. doi: 10.1016/j.jaci.2008.04.039. [DOI] [PubMed] [Google Scholar]
  • 170.Zeyer F., Mothes B., Will C., Carevic M., Rottenberger J., Nürnberg B., Hartl D., Handgretinger R., Beer-Hammer S., Kormann M.S. mRNA-Mediated Gene Supplementation of Toll-Like Receptors as Treatment Strategy for Asthma In Vivo. PLoS ONE. 2016;11:e0154001. doi: 10.1371/journal.pone.0154001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Kreda S.M., Davis C.W., Rose M.C. CFTR, mucins, and mucus obstruction in cystic fibrosis. Cold Spring Harb. Perspect. Med. 2012;2:a009589. doi: 10.1101/cshperspect.a009589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Masood A., Yi M., Belcastro R., Li J., Lopez L., Kantores C., Jankov R.P., Tanswell A.K. Neutrophil elastase-induced elastin degradation mediates macrophage influx and lung injury in 60% O2-exposed neonatal rats. Am. J. Physiol. Lung Cell. Mol. Physiol. 2015;309:L53–L62. doi: 10.1152/ajplung.00298.2014. [DOI] [PubMed] [Google Scholar]
  • 173.Du M., Liu X., Welch E.M., Hirawat S., Peltz S.W., Bedwell D.M. PTC124 is an orally bioavailable compound that promotes suppression of the human CFTR-G542X nonsense allele in a CF mouse model. Proc. Natl. Acad. Sci. USA. 2008;105:2064–2069. doi: 10.1073/pnas.0711795105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Aslam A.A., Higgins C., Sinha I.P., Southern K.W. Ataluren and similar compounds (specific therapies for premature termination codon class I mutations) for cystic fibrosis. Cochrane Database Syst. Rev. 2017;1:CD012040. doi: 10.1002/14651858.CD012040.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Mayer M. Lumacaftor-ivacaftor (Orkambi) for cystic fibrosis: behind the ‘breakthrough’. Evid. Based Med. 2016;21:83–86. doi: 10.1136/ebmed-2015-110325. [DOI] [PubMed] [Google Scholar]
  • 176.Rowe S.M., Daines C., Ringshausen F.C., Kerem E., Wilson J., Tullis E., Nair N., Simard C., Han L., Ingenito E.P. Tezacaftor-Ivacaftor in Residual-Function Heterozygotes with Cystic Fibrosis. N. Engl. J. Med. 2017;377:2024–2035. doi: 10.1056/NEJMoa1709847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Sermet-Gaudelus I., Clancy J.P., Nichols D.P., Nick J.A., De Boeck K., Solomon G.M., Mall M.A., Bolognese J., Bouisset F., den Hollander W. Antisense oligonucleotide eluforsen improves CFTR function in F508del cystic fibrosis. J. Cyst. Fibros. 2018 doi: 10.1016/j.jcf.2018.10.015. Published online November 19, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Ramjeesingh M., Huan L.J., Wilschanski M., Durie P., Li C., Gyomorey K., Wang Y., Kent G., Tanswell K.A., Cutz E. Assessment of the efficacy of in vivo CFTR protein replacement therapy in CF mice. Hum. Gene Ther. 1998;9:521–528. doi: 10.1089/hum.1998.9.4-521. [DOI] [PubMed] [Google Scholar]
  • 179.Alton E.W.F.W., Armstrong D.K., Ashby D., Bayfield K.J., Bilton D., Bloomfield E.V., Boyd A.C., Brand J., Buchan R., Calcedo R., UK Cystic Fibrosis Gene Therapy Consortium Repeated nebulisation of non-viral CFTR gene therapy in patients with cystic fibrosis: a randomised, double-blind, placebo-controlled, phase 2b trial. Lancet Respir. Med. 2015;3:684–691. doi: 10.1016/S2213-2600(15)00245-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Alton E.W.F.W., Armstrong D.K., Ashby D., Bayfield K.J., Bilton D., Bloomfield E.V., Boyd A.C., Brand J., Buchan R., Calcedo R. A randomised, double-blind, placebo-controlled trial of repeated nebulisation of non-viral cystic fibrosis transmembrane conductance regulator (CFTR) gene therapy in patients with cystic fibrosis. Efficacy and Mechanism Evaluation. 2016;3:1–240. [PubMed] [Google Scholar]
  • 181.Robinson E., MacDonald K.D., Slaughter K., McKinney M., Patel S., Sun C., Sahay G. Lipid Nanoparticle-Delivered Chemically Modified mRNA Restores Chloride Secretion in Cystic Fibrosis. Mol. Ther. 2018;26:2034–2046. doi: 10.1016/j.ymthe.2018.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Bangel-Ruland N., Tomczak K., Fernández Fernández E., Leier G., Leciejewski B., Rudolph C., Rosenecker J., Weber W.M. Cystic fibrosis transmembrane conductance regulator-mRNA delivery: a novel alternative for cystic fibrosis gene therapy. J. Gene Med. 2013;15:414–426. doi: 10.1002/jgm.2748. [DOI] [PubMed] [Google Scholar]
  • 183.Suzuki M., Sze M.A., Campbell J.D., Brothers J.F., 2nd, Lenburg M.E., McDonough J.E., Elliott W.M., Cooper J.D., Spira A., Hogg J.C. The cellular and molecular determinants of emphysematous destruction in COPD. Sci. Rep. 2017;7:9562. doi: 10.1038/s41598-017-10126-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Laurell C.B., Eriksson S. The electrophoretic α1-globulin pattern of serum in α1-antitrypsin deficiency. 1963. COPD. 2013;10(Suppl 1):3–8. doi: 10.3109/15412555.2013.771956. [DOI] [PubMed] [Google Scholar]
  • 185.Gadek J.E., Fells G.A., Zimmerman R.L., Rennard S.I., Crystal R.G. Antielastases of the human alveolar structures. Implications for the protease-antiprotease theory of emphysema. J. Clin. Invest. 1981;68:889–898. doi: 10.1172/JCI110344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Lomas D.A., Evans D.L., Finch J.T., Carrell R.W. The mechanism of Z alpha 1-antitrypsin accumulation in the liver. Nature. 1992;357:605–607. doi: 10.1038/357605a0. [DOI] [PubMed] [Google Scholar]
  • 187.Escribano A., Amor M., Pastor S., Castillo S., Sanz F., Codoñer-Franch P., Dasí F. Decreased glutathione and low catalase activity contribute to oxidative stress in children with α-1 antitrypsin deficiency. Thorax. 2015;70:82–83. doi: 10.1136/thoraxjnl-2014-205898. [DOI] [PubMed] [Google Scholar]
  • 188.Escribano A., Pastor S., Reula A., Castillo S., Vicente S., Sanz F., Casas F., Torres M., Fernández-Fabrellas E., Codoñer-Franch P., Dasí F. Accelerated telomere attrition in children and teenagers with α1-antitrypsin deficiency. Eur. Respir. J. 2016;48:350–358. doi: 10.1183/13993003.00176-2016. [DOI] [PubMed] [Google Scholar]
  • 189.Alam S., Li Z., Janciauskiene S., Mahadeva R. Oxidation of Z α1-antitrypsin by cigarette smoke induces polymerization: a novel mechanism of early-onset emphysema. Am. J. Respir. Cell Mol. Biol. 2011;45:261–269. doi: 10.1165/rcmb.2010-0328OC. [DOI] [PubMed] [Google Scholar]
  • 190.Ronzoni R., Berardelli R., Medicina D., Sitia R., Gooptu B., Fra A.M. Aberrant disulphide bonding contributes to the ER retention of alpha1-antitrypsin deficiency variants. Hum. Mol. Genet. 2016;25:642–650. doi: 10.1093/hmg/ddv501. [DOI] [PubMed] [Google Scholar]
  • 191.Pittschieler K. Vitamin E and liver damage in MZ heterozygous infants with alpha 1-antitrypsin deficiency. Acta Paediatr. 1993;82:228–232. doi: 10.1111/j.1651-2227.1993.tb12647.x. [DOI] [PubMed] [Google Scholar]
  • 192.Kode A., Rajendrasozhan S., Caito S., Yang S.R., Megson I.L., Rahman I. Resveratrol induces glutathione synthesis by activation of Nrf2 and protects against cigarette smoke-mediated oxidative stress in human lung epithelial cells. Am. J. Physiol. Lung Cell. Mol. Physiol. 2008;294:L478–L488. doi: 10.1152/ajplung.00361.2007. [DOI] [PubMed] [Google Scholar]
  • 193.Novoradovsky A., Brantly M.L., Waclawiw M.A., Chaudhary P.P., Ihara H., Qi L., Eissa N.T., Barnes P.M., Gabriele K.M., Ehrmantraut M.E. Endothelial nitric oxide synthase as a potential susceptibility gene in the pathogenesis of emphysema in alpha1-antitrypsin deficiency. Am. J. Respir. Cell Mol. Biol. 1999;20:441–447. doi: 10.1165/ajrcmb.20.3.3144. [DOI] [PubMed] [Google Scholar]
  • 194.Topic A., Nagorni-Obradovic L., Francuski D., Ljujic M., Malic Z., Radojkovic D. Oxidative Stress and Polymorphism of Xenobiotic-Metabolizing Enzymes in Two Patients with Severe Alpha-1-Antitrypsin Deficiency. Biochem. Genet. 2016;54:746–752. doi: 10.1007/s10528-016-9748-7. [DOI] [PubMed] [Google Scholar]
  • 195.Wood A.M., Simmonds M.J., Bayley D.L., Newby P.R., Gough S.C., Stockley R.A. The TNFalpha gene relates to clinical phenotype in alpha-1-antitrypsin deficiency. Respir. Res. 2008;9:52. doi: 10.1186/1465-9921-9-52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Kim W.J., Wood A.M., Barker A.F., Brantly M.L., Campbell E.J., Eden E., McElvaney G., Rennard S.I., Sandhaus R.A., Stocks J.M. Association of IREB2 and CHRNA3 polymorphisms with airflow obstruction in severe alpha-1 antitrypsin deficiency. Respir. Res. 2012;13:16. doi: 10.1186/1465-9921-13-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Wewers M.D., Gadek J.E., Keogh B.A., Fells G.A., Crystal R.G. Evaluation of danazol therapy for patients with PiZZ alpha-1-antitrypsin deficiency. Am. Rev. Respir. Dis. 1986;134:476–480. doi: 10.1164/arrd.1986.134.3.476. [DOI] [PubMed] [Google Scholar]
  • 198.Mallya M., Phillips R.L., Saldanha S.A., Gooptu B., Brown S.C., Termine D.J., Shirvani A.M., Wu Y., Sifers R.N., Abagyan R., Lomas D.A. Small molecules block the polymerization of Z alpha1-antitrypsin and increase the clearance of intracellular aggregates. J. Med. Chem. 2007;50:5357–5363. doi: 10.1021/jm070687z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Mahadeva R., Dafforn T.R., Carrell R.W., Lomas D.A. 6-mer peptide selectively anneals to a pathogenic serpin conformation and blocks polymerization. Implications for the prevention of Z alpha(1)-antitrypsin-related cirrhosis. J. Biol. Chem. 2002;277:6771–6774. doi: 10.1074/jbc.C100722200. [DOI] [PubMed] [Google Scholar]
  • 200.Hidvegi T., Ewing M., Hale P., Dippold C., Beckett C., Kemp C., Maurice N., Mukherjee A., Goldbach C., Watkins S. An autophagy-enhancing drug promotes degradation of mutant alpha1-antitrypsin Z and reduces hepatic fibrosis. Science. 2010;329:229–232. doi: 10.1126/science.1190354. [DOI] [PubMed] [Google Scholar]
  • 201.Maurice N., Perlmutter D.H. Novel treatment strategies for liver disease due to α1-antitrypsin deficiency. Clin. Transl. Sci. 2012;5:289–294. doi: 10.1111/j.1752-8062.2011.00363.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Kaushal S., Annamali M., Blomenkamp K., Rudnick D., Halloran D., Brunt E.M., Teckman J.H. Rapamycin reduces intrahepatic alpha-1-antitrypsin mutant Z protein polymers and liver injury in a mouse model. Exp. Biol. Med. (Maywood) 2010;235:700–709. doi: 10.1258/ebm.2010.009297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Hassan T., Carroll T.P., Buckley P.G., Cummins R., O’Neill S.J., McElvaney N.G., Greene C.M. miR-199a-5p silencing regulates the unfolded protein response in chronic obstructive pulmonary disease and α1-antitrypsin deficiency. Am. J. Respir. Crit. Care Med. 2014;189:263–273. doi: 10.1164/rccm.201306-1151OC. [DOI] [PubMed] [Google Scholar]
  • 204.Nana-Sinkam S.P., Choi A.M. Epigenetics and the unfolded protein response in the lung: emerging role for microRNAs. Am. J. Respir. Crit. Care Med. 2014;189:239–240. doi: 10.1164/rccm.201312-2230ED. [DOI] [PubMed] [Google Scholar]
  • 205.Esquinas C., Janciauskiene S., Gonzalo R., Mas de Xaxars G., Olejnicka B., Belmonte I., Barrecheguren M., Rodriguez E., Nuñez A., Rodriguez-Frias F., Miravitlles M. Gene and miRNA expression profiles in PBMCs from patients with severe and mild emphysema and PiZZ alpha1-antitrypsin deficiency. Int. J. Chron. Obstruct. Pulmon. Dis. 2017;12:3381–3390. doi: 10.2147/COPD.S145445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Stoller J.K., Rouhani F., Brantly M., Shahin S., Dweik R.A., Stocks J.M., Clausen J., Campbell E., Norton F. Biochemical efficacy and safety of a new pooled human plasma alpha(1)-antitrypsin, Respitin. Chest. 2002;122:66–74. doi: 10.1378/chest.122.1.66. [DOI] [PubMed] [Google Scholar]
  • 207.Sandhaus R.A. Alpha 1-antitrypsin augmentation therapy. Agents Actions Suppl. 1993;42:97–102. [PubMed] [Google Scholar]
  • 208.Chapman K.R., Burdon J.G., Piitulainen E., Sandhaus R.A., Seersholm N., Stocks J.M., Stoel B.C., Huang L., Yao Z., Edelman J.M., McElvaney N.G., RAPID Trial Study Group Intravenous augmentation treatment and lung density in severe α1 antitrypsin deficiency (RAPID): a randomised, double-blind, placebo-controlled trial. Lancet. 2015;386:360–368. doi: 10.1016/S0140-6736(15)60860-1. [DOI] [PubMed] [Google Scholar]
  • 209.Chiuchiolo M.J., Crystal R.G. Gene Therapy for Alpha-1 Antitrypsin Deficiency Lung Disease. Ann. Am. Thorac. Soc. 2016;13(Suppl 4):S352–S369. doi: 10.1513/AnnalsATS.201506-344KV. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Chiuchiolo M.J., Kaminsky S.M., Sondhi D., Mancenido D., Hollmann C., Crystal R.G. Phase I/II study of intrapleural administration of a serotype rh.10 replication-deficient adeno-associated virus gene transfer vector expressing the human α1-antitrypsin cDNA to individuals with α1-antitrypsin deficiency. Hum. Gene Ther. Clin. Dev. 2014;25:112–133. doi: 10.1089/humc.2014.2513. [DOI] [PubMed] [Google Scholar]
  • 211.Michel T., Kankura A., Salinas Medina M.L., Kurz J., Behring A., Avci-Adali M., Nolte A., Schlensak C., Wendel H.P., Krajewski S. In Vitro Evaluation of a Novel mRNA-Based Therapeutic Strategy for the Treatment of Patients Suffering from Alpha-1-Antitrypsin Deficiency. Nucleic Acid Ther. 2015;25:235–244. doi: 10.1089/nat.2015.0537. [DOI] [PubMed] [Google Scholar]
  • 212.Karikó K., Kuo A., Barnathan E. Overexpression of urokinase receptor in mammalian cells following administration of the in vitro transcribed encoding mRNA. Gene Ther. 1999;6:1092–1100. doi: 10.1038/sj.gt.3300930. [DOI] [PubMed] [Google Scholar]
  • 213.Kallen K.J., Theß A. A development that may evolve into a revolution in medicine: mRNA as the basis for novel, nucleotide-based vaccines and drugs. Ther. Adv. Vaccines. 2014;2:10–31. doi: 10.1177/2051013613508729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Svitkin Y.V., Cheng Y.M., Chakraborty T., Presnyak V., John M., Sonenberg N. N1-methyl-pseudouridine in mRNA enhances translation through eIF2α-dependent and independent mechanisms by increasing ribosome density. Nucleic Acids Res. 2017;45:6023–6036. doi: 10.1093/nar/gkx135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Vaidyanathan S., Azizian K.T., Haque A.K.M.A., Henderson J.M., Hendel A., Shore S., Antony J.S., Hogrefe R.I., Kormann M.S.D., Porteus M.H., McCaffrey A.P. Uridine Depletion and Chemical Modification Increase Cas9 mRNA Activity and Reduce Immunogenicity without HPLC Purification. Mol. Ther. Nucleic Acids. 2018;12:530–542. doi: 10.1016/j.omtn.2018.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Filipowicz W., Furuichi Y., Sierra J.M., Muthukrishnan S., Shatkin A.J., Ochoa S. A protein binding the methylated 5′-terminal sequence, m7GpppN, of eukaryotic messenger RNA. Proc. Natl. Acad. Sci. USA. 1976;73:1559–1563. doi: 10.1073/pnas.73.5.1559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.van Dijk E., Cougot N., Meyer S., Babajko S., Wahle E., Séraphin B. Human Dcp2: a catalytically active mRNA decapping enzyme located in specific cytoplasmic structures. EMBO J. 2002;21:6915–6924. doi: 10.1093/emboj/cdf678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Liu H., Rodgers N.D., Jiao X., Kiledjian M. The scavenger mRNA decapping enzyme DcpS is a member of the HIT family of pyrophosphatases. EMBO J. 2002;21:4699–4708. doi: 10.1093/emboj/cdf448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Mazza C., Segref A., Mattaj I.W., Cusack S. Large-scale induced fit recognition of an m(7)GpppG cap analogue by the human nuclear cap-binding complex. EMBO J. 2002;21:5548–5557. doi: 10.1093/emboj/cdf538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Stepinski J., Waddell C., Stolarski R., Darzynkiewicz E., Rhoads R.E. Synthesis and properties of mRNAs containing the novel “anti-reverse” cap analogs 7-methyl(3′-O-methyl)GpppG and 7-methyl (3′-deoxy)GpppG. RNA. 2001;7:1486–1495. [PMC free article] [PubMed] [Google Scholar]
  • 221.Furuichi Y., Shatkin A.J. Viral and cellular mRNA capping: past and prospects. Adv. Virus Res. 2000;55:135–184. doi: 10.1016/S0065-3527(00)55003-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Katibah G.E., Qin Y., Sidote D.J., Yao J., Lambowitz A.M., Collins K. Broad and adaptable RNA structure recognition by the human interferon-induced tetratricopeptide repeat protein IFIT5. Proc. Natl. Acad. Sci. USA. 2014;111:12025–12030. doi: 10.1073/pnas.1412842111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Abbas Y.M., Laudenbach B.T., Martínez-Montero S., Cencic R., Habjan M., Pichlmair A., Damha M.J., Pelletier J., Nagar B. Structure of human IFIT1 with capped RNA reveals adaptable mRNA binding and mechanisms for sensing N1 and N2 ribose 2′-O methylations. Proc. Natl. Acad. Sci. USA. 2017;114:E2106–E2115. doi: 10.1073/pnas.1612444114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Schoggins J.W. A Sense of Self: RIG-I’s Tolerance to Host RNA. Immunity. 2015;43:1–2. doi: 10.1016/j.immuni.2015.06.022. [DOI] [PubMed] [Google Scholar]
  • 225.Körner C.G., Wahle E. Poly(A) tail shortening by a mammalian poly(A)-specific 3′-exoribonuclease. J. Biol. Chem. 1997;272:10448–10456. doi: 10.1074/jbc.272.16.10448. [DOI] [PubMed] [Google Scholar]
  • 226.Quabius E.S., Krupp G. Synthetic mRNAs for manipulating cellular phenotypes: an overview. N. Biotechnol. 2015;32:229–235. doi: 10.1016/j.nbt.2014.04.008. [DOI] [PubMed] [Google Scholar]
  • 227.Bergman N., Moraes K.C., Anderson J.R., Zaric B., Kambach C., Schneider R.J., Wilusz C.J., Wilusz J. Lsm proteins bind and stabilize RNAs containing 5′ poly(A) tracts. Nat. Struct. Mol. Biol. 2007;14:824–831. doi: 10.1038/nsmb1287. [DOI] [PubMed] [Google Scholar]
  • 228.Chen C.Y., Shyu A.B. AU-rich elements: characterization and importance in mRNA degradation. Trends Biochem. Sci. 1995;20:465–470. doi: 10.1016/s0968-0004(00)89102-1. [DOI] [PubMed] [Google Scholar]
  • 229.Machnicka M.A., Milanowska K., Osman Oglou O., Purta E., Kurkowska M., Olchowik A., Januszewski W., Kalinowski S., Dunin-Horkawicz S., Rother K.M. MODOMICS: a database of RNA modification pathways--2013 update. Nucleic Acids Res. 2013;41:D262–D267. doi: 10.1093/nar/gks1007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Karikó K., Buckstein M., Ni H., Weissman D. Suppression of RNA recognition by Toll-like receptors: the impact of nucleoside modification and the evolutionary origin of RNA. Immunity. 2005;23:165–175. doi: 10.1016/j.immuni.2005.06.008. [DOI] [PubMed] [Google Scholar]
  • 231.Karikó K., Muramatsu H., Keller J.M., Weissman D. Increased erythropoiesis in mice injected with submicrogram quantities of pseudouridine-containing mRNA encoding erythropoietin. Mol. Ther. 2012;20:948–953. doi: 10.1038/mt.2012.7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Huang H., Weng H., Sun W., Qin X., Shi H., Wu H., Zhao B.S., Mesquita A., Liu C., Yuan C.L. Recognition of RNA N6-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nat. Cell Biol. 2018;20:285–295. doi: 10.1038/s41556-018-0045-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Warren L., Manos P.D., Ahfeldt T., Loh Y.H., Li H., Lau F., Ebina W., Mandal P.K., Smith Z.D., Meissner A. Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA. Cell Stem Cell. 2010;7:618–630. doi: 10.1016/j.stem.2010.08.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Durbin A.F., Wang C., Marcotrigiano J., Gehrke L. RNAs Containing Modified Nucleotides Fail To Trigger RIG-I Conformational Changes for Innate Immune Signaling. MBio. 2016;7:e00833-16. doi: 10.1128/mBio.00833-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Anderson B.R., Muramatsu H., Nallagatla S.R., Bevilacqua P.C., Sansing L.H., Weissman D., Karikó K. Incorporation of pseudouridine into mRNA enhances translation by diminishing PKR activation. Nucleic Acids Res. 2010;38:5884–5892. doi: 10.1093/nar/gkq347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Yang X., Yang Y., Sun B.-F., Chen Y.-S., Xu J.-W., Lai W.-Y., Li A., Wang X., Bhattarai D.P., Xiao W. 5-methylcytosine promotes mRNA export - NSUN2 as the methyltransferase and ALYREF as an m5C reader. Cell Res. 2017;27:606–625. doi: 10.1038/cr.2017.55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Anderson B.R., Muramatsu H., Jha B.K., Silverman R.H., Weissman D., Karikó K. Nucleoside modifications in RNA limit activation of 2′-5′-oligoadenylate synthetase and increase resistance to cleavage by RNase L. Nucleic Acids Res. 2011;39:9329–9338. doi: 10.1093/nar/gkr586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Andries O., Mc Cafferty S., De Smedt S.C., Weiss R., Sanders N.N., Kitada T. N(1)-methylpseudouridine-incorporated mRNA outperforms pseudouridine-incorporated mRNA by providing enhanced protein expression and reduced immunogenicity in mammalian cell lines and mice. J. Control. Release. 2015;217:337–344. doi: 10.1016/j.jconrel.2015.08.051. [DOI] [PubMed] [Google Scholar]
  • 239.Pardi N., Tuyishime S., Muramatsu H., Kariko K., Mui B.L., Tam Y.K., Madden T.D., Hope M.J., Weissman D. Expression kinetics of nucleoside-modified mRNA delivered in lipid nanoparticles to mice by various routes. J. Control. Release. 2015;217:345–351. doi: 10.1016/j.jconrel.2015.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Thess A., Grund S., Mui B.L., Hope M.J., Baumhof P., Fotin-Mleczek M., Schlake T. Sequence-engineered mRNA Without Chemical Nucleoside Modifications Enables an Effective Protein Therapy in Large Animals. Mol. Ther. 2015;23:1456–1464. doi: 10.1038/mt.2015.103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Ely L., Roa W., Finlay W.H., Löbenberg R. Effervescent dry powder for respiratory drug delivery. Eur. J. Pharm. Biopharm. 2007;65:346–353. doi: 10.1016/j.ejpb.2006.10.021. [DOI] [PubMed] [Google Scholar]
  • 242.Grenha A., Seijo B., Remuñán-López C. Microencapsulated chitosan nanoparticles for lung protein delivery. Eur. J. Pharm. Sci. 2005;25:427–437. doi: 10.1016/j.ejps.2005.04.009. [DOI] [PubMed] [Google Scholar]
  • 243.Yamamoto H., Kuno Y., Sugimoto S., Takeuchi H., Kawashima Y. Surface-modified PLGA nanosphere with chitosan improved pulmonary delivery of calcitonin by mucoadhesion and opening of the intercellular tight junctions. J. Control. Release. 2005;102:373–381. doi: 10.1016/j.jconrel.2004.10.010. [DOI] [PubMed] [Google Scholar]
  • 244.McConville J.T., Overhoff K.A., Sinswat P., Vaughn J.M., Frei B.L., Burgess D.S., Talbert R.L., Peters J.I., Johnston K.P., Williams R.O., 3rd Targeted high lung concentrations of itraconazole using nebulized dispersions in a murine model. Pharm. Res. 2006;23:901–911. doi: 10.1007/s11095-006-9904-6. [DOI] [PubMed] [Google Scholar]
  • 245.Kreuter J., Täuber U., Illi V. Distribution and elimination of poly(methyl-2-14C-methacrylate) nanoparticle radioactivity after injection in rats and mice. J. Pharm. Sci. 1979;68:1443–1447. doi: 10.1002/jps.2600681129. [DOI] [PubMed] [Google Scholar]
  • 246.Green M.R., Manikhas G.M., Orlov S., Afanasyev B., Makhson A.M., Bhar P., Hawkins M.J. Abraxane, a novel Cremophor-free, albumin-bound particle form of paclitaxel for the treatment of advanced non-small-cell lung cancer. Ann. Oncol. 2006;17:1263–1268. doi: 10.1093/annonc/mdl104. [DOI] [PubMed] [Google Scholar]
  • 247.Kreuter J. Evaluation of nanoparticles as drug-delivery systems. III: materials, stability, toxicity, possibilities of targeting, and use. Pharm. Acta Helv. 1983;58:242–250. [PubMed] [Google Scholar]
  • 248.Shalgunov V., Zaytseva-Zotova D., Zintchenko A., Levada T., Shilov Y., Andreyev D., Dzhumashev D., Metelkin E., Urusova A., Demin O. Comprehensive study of the drug delivery properties of poly(l-lactide)-poly(ethylene glycol) nanoparticles in rats and tumor-bearing mice. J. Control. Release. 2017;261:31–42. doi: 10.1016/j.jconrel.2017.06.006. [DOI] [PubMed] [Google Scholar]
  • 249.Wang T., Upponi J.R., Torchilin V.P. Design of multifunctional non-viral gene vectors to overcome physiological barriers: dilemmas and strategies. Int. J. Pharm. 2012;427:3–20. doi: 10.1016/j.ijpharm.2011.07.013. [DOI] [PubMed] [Google Scholar]
  • 250.Sahin U., Karikó K., Türeci Ö. mRNA-based therapeutics--developing a new class of drugs. Nat. Rev. Drug Discov. 2014;13:759–780. doi: 10.1038/nrd4278. [DOI] [PubMed] [Google Scholar]
  • 251.Sanders N., Rudolph C., Braeckmans K., De Smedt S.C., Demeester J. Extracellular barriers in respiratory gene therapy. Adv. Drug Deliv. Rev. 2009;61:115–127. doi: 10.1016/j.addr.2008.09.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Bosquillon C., Lombry C., Préat V., Vanbever R. Influence of formulation excipients and physical characteristics of inhalation dry powders on their aerosolization performance. J. Control. Release. 2001;70:329–339. doi: 10.1016/s0168-3659(00)00362-x. [DOI] [PubMed] [Google Scholar]
  • 253.Newman S.P. Aerosol deposition considerations in inhalation therapy. Chest. 1985;88(2, Suppl):152S–160S. doi: 10.1378/chest.88.2_supplement.152s. [DOI] [PubMed] [Google Scholar]
  • 254.Wine J.J. The genesis of cystic fibrosis lung disease. J. Clin. Invest. 1999;103:309–312. doi: 10.1172/JCI6222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Widdicombe J.G. Airway liquid: a barrier to drug diffusion? Eur. Respir. J. 1997;10:2194–2197. doi: 10.1183/09031936.97.10102194. [DOI] [PubMed] [Google Scholar]
  • 256.Button B., Cai L.H., Ehre C., Kesimer M., Hill D.B., Sheehan J.K., Boucher R.C., Rubinstein M. A periciliary brush promotes the lung health by separating the mucus layer from airway epithelia. Science. 2012;337:937–941. doi: 10.1126/science.1223012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Yeates D.B., Aspin N., Levison H., Jones M.T., Bryan A.C. Mucociliary tracheal transport rates in man. J. Appl. Physiol. 1975;39:487–495. doi: 10.1152/jappl.1975.39.3.487. [DOI] [PubMed] [Google Scholar]
  • 258.Sanders N.N., De Smedt S.C., Van Rompaey E., Simoens P., De Baets F., Demeester J. Cystic fibrosis sputum: a barrier to the transport of nanospheres. Am. J. Respir. Crit. Care Med. 2000;162:1905–1911. doi: 10.1164/ajrccm.162.5.9909009. [DOI] [PubMed] [Google Scholar]
  • 259.Dawson M., Wirtz D., Hanes J. Enhanced viscoelasticity of human cystic fibrotic sputum correlates with increasing microheterogeneity in particle transport. J. Biol. Chem. 2003;278:50393–50401. doi: 10.1074/jbc.M309026200. [DOI] [PubMed] [Google Scholar]
  • 260.Broughton-Head V.J., Smith J.R., Shur J., Shute J.K. Actin limits enhancement of nanoparticle diffusion through cystic fibrosis sputum by mucolytics. Pulm. Pharmacol. Ther. 2007;20:708–717. doi: 10.1016/j.pupt.2006.08.008. [DOI] [PubMed] [Google Scholar]
  • 261.Stern M., Caplen N.J., Browning J.E., Griesenbach U., Sorgi F., Huang L., Gruenert D.C., Marriot C., Crystal R.G., Geddes D.M., Alton E.W. The effect of mucolytic agents on gene transfer across a CF sputum barrier in vitro. Gene Ther. 1998;5:91–98. doi: 10.1038/sj.gt.3300556. [DOI] [PubMed] [Google Scholar]
  • 262.Kitson C., Angel B., Judd D., Rothery S., Severs N.J., Dewar A., Huang L., Wadsworth S.C., Cheng S.H., Geddes D.M., Alton E.W. The extra- and intracellular barriers to lipid and adenovirus-mediated pulmonary gene transfer in native sheep airway epithelium. Gene Ther. 1999;6:534–546. doi: 10.1038/sj.gt.3300840. [DOI] [PubMed] [Google Scholar]
  • 263.Ferrari S., Kitson C., Farley R., Steel R., Marriott C., Parkins D.A., Scarpa M., Wainwright B., Evans M.J., Colledge W.H. Mucus altering agents as adjuncts for nonviral gene transfer to airway epithelium. Gene Ther. 2001;8:1380–1386. doi: 10.1038/sj.gt.3301525. [DOI] [PubMed] [Google Scholar]
  • 264.Ernst N., Ulrichskötter S., Schmalix W.A., Rädler J., Galneder R., Mayer E., Gersting S., Plank C., Reinhardt D., Rosenecker J. Interaction of liposomal and polycationic transfection complexes with pulmonary surfactant. J. Gene Med. 1999;1:331–340. doi: 10.1002/(SICI)1521-2254(199909/10)1:5<331::AID-JGM60>3.0.CO;2-8. [DOI] [PubMed] [Google Scholar]
  • 265.Kukowska-Latallo J.F., Chen C., Eichman J., Bielinska A.U., Baker J.R., Jr. Enhancement of dendrimer-mediated transfection using synthetic lung surfactant exosurf neonatal in vitro. Biochem. Biophys. Res. Commun. 1999;264:253–261. doi: 10.1006/bbrc.1999.1458. [DOI] [PubMed] [Google Scholar]
  • 266.Sanders N.N., Van Rompaey E., De Smedt S.C., Demeester J. On the transport of lipoplexes through cystic fibrosis sputum. Pharm. Res. 2002;19:451–456. doi: 10.1023/a:1015139527747. [DOI] [PubMed] [Google Scholar]
  • 267.King M., Rubin B.K. Pharmacological approaches to discovery and development of new mucolytic agents. Adv. Drug Deliv. Rev. 2002;54:1475–1490. doi: 10.1016/s0169-409x(02)00156-4. [DOI] [PubMed] [Google Scholar]
  • 268.Rubin B.K. Mucolytics, expectorants, and mucokinetic medications. Respir. Care. 2007;52:859–865. [PubMed] [Google Scholar]
  • 269.Sanders N.N., De Smedt S.C., Cheng S.H., Demeester J. Pegylated GL67 lipoplexes retain their gene transfection activity after exposure to components of CF mucus. Gene Ther. 2002;9:363–371. doi: 10.1038/sj.gt.3301663. [DOI] [PubMed] [Google Scholar]
  • 270.Maisel K., Reddy M., Xu Q., Chattopadhyay S., Cone R., Ensign L.M., Hanes J. Nanoparticles coated with high molecular weight PEG penetrate mucus and provide uniform vaginal and colorectal distribution in vivo. Nanomedicine (Lond.) 2016;11:1337–1343. doi: 10.2217/nnm-2016-0047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Neumann E., Schaefer-Ridder M., Wang Y., Hofschneider P.H. Gene transfer into mouse lyoma cells by electroporation in high electric fields. EMBO J. 1982;1:841–845. doi: 10.1002/j.1460-2075.1982.tb01257.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Mattozzi M.D., Voges M.J., Silver P.A., Way J.C. Transient gene expression in tobacco using Gibson assembly and the Gene Gun. J. Vis. Exp. 2014;(86):51234. doi: 10.3791/51234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Celis J.E. Expression of mRNAs microinjected into somatic cells. Ciba Found. Symp. 1984;103:220–238. doi: 10.1002/9780470720844.ch14. [DOI] [PubMed] [Google Scholar]
  • 274.Dewitte H., Van Lint S., Heirman C., Thielemans K., De Smedt S.C., Breckpot K., Lentacker I. The potential of antigen and TriMix sonoporation using mRNA-loaded microbubbles for ultrasound-triggered cancer immunotherapy. J. Control. Release. 2014;194:28–36. doi: 10.1016/j.jconrel.2014.08.011. [DOI] [PubMed] [Google Scholar]
  • 275.Islam M.A., Reesor E.K., Xu Y., Zope H.R., Zetter B.R., Shi J. Biomaterials for mRNA delivery. Biomater. Sci. 2015;3:1519–1533. doi: 10.1039/c5bm00198f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276.Hajj K.A., Whitehead K.A. Tools for translation: non-viral materials for therapeutic mRNA delivery. Nat. Rev. Mater. 2017;2:17056. [Google Scholar]
  • 277.Yin H., Kanasty R.L., Eltoukhy A.A., Vegas A.J., Dorkin J.R., Anderson D.G. Non-viral vectors for gene-based therapy. Nat. Rev. Genet. 2014;15:541–555. doi: 10.1038/nrg3763. [DOI] [PubMed] [Google Scholar]
  • 278.Deering R.P., Kommareddy S., Ulmer J.B., Brito L.A., Geall A.J. Nucleic acid vaccines: prospects for non-viral delivery of mRNA vaccines. Expert Opin. Drug Deliv. 2014;11:885–899. doi: 10.1517/17425247.2014.901308. [DOI] [PubMed] [Google Scholar]
  • 279.Phua K.K., Leong K.W., Nair S.K. Transfection efficiency and transgene expression kinetics of mRNA delivered in naked and nanoparticle format. J. Control. Release. 2013;166:227–233. doi: 10.1016/j.jconrel.2012.12.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280.Khalil I.A., Kogure K., Akita H., Harashima H. Uptake pathways and subsequent intracellular trafficking in nonviral gene delivery. Pharmacol. Rev. 2006;58:32–45. doi: 10.1124/pr.58.1.8. [DOI] [PubMed] [Google Scholar]
  • 281.Lonez C., Vandenbranden M., Ruysschaert J.M. Cationic lipids activate intracellular signaling pathways. Adv. Drug Deliv. Rev. 2012;64:1749–1758. doi: 10.1016/j.addr.2012.05.009. [DOI] [PubMed] [Google Scholar]
  • 282.Noble G.T., Stefanick J.F., Ashley J.D., Kiziltepe T., Bilgicer B. Ligand-targeted liposome design: challenges and fundamental considerations. Trends Biotechnol. 2014;32:32–45. doi: 10.1016/j.tibtech.2013.09.007. [DOI] [PubMed] [Google Scholar]
  • 283.Zangi L., Lui K.O., von Gise A., Ma Q., Ebina W., Ptaszek L.M., Später D., Xu H., Tabebordbar M., Gorbatov R. Modified mRNA directs the fate of heart progenitor cells and induces vascular regeneration after myocardial infarction. Nat. Biotechnol. 2013;31:898–907. doi: 10.1038/nbt.2682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 284.Phua K.K., Staats H.F., Leong K.W., Nair S.K. Intranasal mRNA nanoparticle vaccination induces prophylactic and therapeutic anti-tumor immunity. Sci. Rep. 2014;4:5128. doi: 10.1038/srep05128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285.Antony J.S., Dewerth A., Haque A., Handgretinger R., Kormann M.S. Modified mRNA as a new therapeutic option for pediatric respiratory diseases and hemoglobinopathies. Mol. Cell Pediatr. 2015;2:11. doi: 10.1186/s40348-015-0022-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286.Semple S.C., Akinc A., Chen J., Sandhu A.P., Mui B.L., Cho C.K., Sah D.W., Stebbing D., Crosley E.J., Yaworski E. Rational design of cationic lipids for siRNA delivery. Nat. Biotechnol. 2010;28:172–176. doi: 10.1038/nbt.1602. [DOI] [PubMed] [Google Scholar]
  • 287.Litzinger D.C., Brown J.M., Wala I., Kaufman S.A., Van G.Y., Farrell C.L., Collins D. Fate of cationic liposomes and their complex with oligonucleotide in vivo. Biochim. Biophys. Acta. 1996;1281:139–149. doi: 10.1016/0005-2736(95)00268-5. [DOI] [PubMed] [Google Scholar]
  • 288.Ma Z., Zhang J., Alber S., Dileo J., Negishi Y., Stolz D., Watkins S., Huang L., Pitt B., Li S. Lipid-mediated delivery of oligonucleotide to pulmonary endothelium. Am. J. Respir. Cell Mol. Biol. 2002;27:151–159. doi: 10.1165/ajrcmb.27.2.4653. [DOI] [PubMed] [Google Scholar]
  • 289.Eastman S.J., Lukason M.J., Tousignant J.D., Murray H., Lane M.D., St George J.A., Akita G.Y., Cherry M., Cheng S.H., Scheule R.K. A concentrated and stable aerosol formulation of cationic lipid:DNA complexes giving high-level gene expression in mouse lung. Hum. Gene Ther. 1997;8:765–773. doi: 10.1089/hum.1997.8.6-765. [DOI] [PubMed] [Google Scholar]
  • 290.Lee E.R., Marshall J., Siegel C.S., Jiang C., Yew N.S., Nichols M.R., Nietupski J.B., Ziegler R.J., Lane M.B., Wang K.X. Detailed analysis of structures and formulations of cationic lipids for efficient gene transfer to the lung. Hum. Gene Ther. 1996;7:1701–1717. doi: 10.1089/hum.1996.7.14-1701. [DOI] [PubMed] [Google Scholar]
  • 291.Scheule R.K., St George J.A., Bagley R.G., Marshall J., Kaplan J.M., Akita G.Y., Wang K.X., Lee E.R., Harris D.J., Jiang C. Basis of pulmonary toxicity associated with cationic lipid-mediated gene transfer to the mammalian lung. Hum. Gene Ther. 1997;8:689–707. doi: 10.1089/hum.1997.8.6-689. [DOI] [PubMed] [Google Scholar]
  • 292.Michel T., Luft D., Abraham M.K., Reinhardt S., Salinas Medina M.L., Kurz J., Schaller M., Avci-Adali M., Schlensak C., Peter K. Cationic Nanoliposomes Meet mRNA: Efficient Delivery of Modified mRNA Using Hemocompatible and Stable Vectors for Therapeutic Applications. Mol. Ther. Nucleic Acids. 2017;8:459–468. doi: 10.1016/j.omtn.2017.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 293.Borel F., Sun H., Zieger M., Cox A., Cardozo B., Li W., Oliveira G., Davis A., Gruntman A., Flotte T.R. Editing out five Serpina1 paralogs to create a mouse model of genetic emphysema. Proc. Natl. Acad. Sci. USA. 2018;115:2788–2793. doi: 10.1073/pnas.1713689115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 294.Ramaswamy S., Tonnu N., Tachikawa K., Limphong P., Vega J.B., Karmali P.P., Chivukula P., Verma I.M. Systemic delivery of factor IX messenger RNA for protein replacement therapy. Proc. Natl. Acad. Sci. USA. 2017;114:E1941–E1950. doi: 10.1073/pnas.1619653114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295.Akinc A., Thomas M., Klibanov A.M., Langer R. Exploring polyethylenimine-mediated DNA transfection and the proton sponge hypothesis. J. Gene Med. 2005;7:657–663. doi: 10.1002/jgm.696. [DOI] [PubMed] [Google Scholar]
  • 296.Boussif O., Lezoualc’h F., Zanta M.A., Mergny M.D., Scherman D., Demeneix B., Behr J.P. A versatile vector for gene and oligonucleotide transfer into cells in culture and in vivo: polyethylenimine. Proc. Natl. Acad. Sci. USA. 1995;92:7297–7301. doi: 10.1073/pnas.92.16.7297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 297.Lungwitz U., Breunig M., Blunk T., Göpferich A. Polyethylenimine-based non-viral gene delivery systems. Eur. J. Pharm. Biopharm. 2005;60:247–266. doi: 10.1016/j.ejpb.2004.11.011. [DOI] [PubMed] [Google Scholar]
  • 298.Lv H., Zhang S., Wang B., Cui S., Yan J. Toxicity of cationic lipids and cationic polymers in gene delivery. J. Control. Release. 2006;114:100–109. doi: 10.1016/j.jconrel.2006.04.014. [DOI] [PubMed] [Google Scholar]
  • 299.Schaffer D.V., Fidelman N.A., Dan N., Lauffenburger D.A. Vector unpacking as a potential barrier for receptor-mediated polyplex gene delivery. Biotechnol. Bioeng. 2000;67:598–606. doi: 10.1002/(sici)1097-0290(20000305)67:5<598::aid-bit10>3.0.co;2-g. [DOI] [PubMed] [Google Scholar]
  • 300.Zhao M., Li M., Zhang Z., Gong T., Sun X. Induction of HIV-1 gag specific immune responses by cationic micelles mediated delivery of gag mRNA. Drug Deliv. 2016;23:2596–2607. doi: 10.3109/10717544.2015.1038856. [DOI] [PubMed] [Google Scholar]
  • 301.Convertine A.J., Benoit D.S., Duvall C.L., Hoffman A.S., Stayton P.S. Development of a novel endosomolytic diblock copolymer for siRNA delivery. J. Control. Release. 2009;133:221–229. doi: 10.1016/j.jconrel.2008.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302.Cheng C., Convertine A.J., Stayton P.S., Bryers J.D. Multifunctional triblock copolymers for intracellular messenger RNA delivery. Biomaterials. 2012;33:6868–6876. doi: 10.1016/j.biomaterials.2012.06.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 303.Suk J.S., Lai S.K., Wang Y.Y., Ensign L.M., Zeitlin P.L., Boyle M.P., Hanes J. The penetration of fresh undiluted sputum expectorated by cystic fibrosis patients by non-adhesive polymer nanoparticles. Biomaterials. 2009;30:2591–2597. doi: 10.1016/j.biomaterials.2008.12.076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 304.Schuster B.S., Suk J.S., Woodworth G.F., Hanes J. Nanoparticle diffusion in respiratory mucus from humans without lung disease. Biomaterials. 2013;34:3439–3446. doi: 10.1016/j.biomaterials.2013.01.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 305.Schneider C.S., Xu Q., Boylan N.J., Chisholm J., Tang B.C., Schuster B.S., Henning A., Ensign L.M., Lee E., Adstamongkonkul P. Nanoparticles that do not adhere to mucus provide uniform and long-lasting drug delivery to airways following inhalation. Sci. Adv. 2017;3:e1601556. doi: 10.1126/sciadv.1601556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 306.Mahiny A.J., Dewerth A., Mays L.E., Alkhaled M., Mothes B., Malaeksefat E., Loretz B., Rottenberger J., Brosch D.M., Reautschnig P. In vivo genome editing using nuclease-encoding mRNA corrects SP-B deficiency. Nat. Biotechnol. 2015;33:584–586. doi: 10.1038/nbt.3241. [DOI] [PubMed] [Google Scholar]
  • 307.Baba M., Itaka K., Kondo K., Yamasoba T., Kataoka K. Treatment of neurological disorders by introducing mRNA in vivo using polyplex nanomicelles. J. Control. Release. 2015;201:41–48. doi: 10.1016/j.jconrel.2015.01.017. [DOI] [PubMed] [Google Scholar]
  • 308.Matsui A., Uchida S., Ishii T., Itaka K., Kataoka K. Messenger RNA-based therapeutics for the treatment of apoptosis-associated diseases. Sci. Rep. 2015;5:15810. doi: 10.1038/srep15810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 309.Kaczmarek J.C., Patel A.K., Kauffman K.J., Fenton O.S., Webber M.J., Heartlein M.W., DeRosa F., Anderson D.G. Polymer-Lipid Nanoparticles for Systemic Delivery of mRNA to the Lungs. Angew. Chem. Int. Ed. Engl. 2016;55:13808–13812. doi: 10.1002/anie.201608450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 310.Hao Z., Qu B., Wang Y., Tang S., Wang G., Qiu M., Zhang R., Liu Y., Xiao X. Preparation and characterization of lung-targeting ceftiofur-loaded gelatin microspheres. Drug Dev. Ind. Pharm. 2011;37:1422–1428. doi: 10.3109/03639045.2011.584192. [DOI] [PubMed] [Google Scholar]
  • 311.Youngren-Ortiz S.R., Hill D.B., Hoffmann P.R., Morris K.R., Barrett E.G., Forest M.G., Chougule M.B. Development of Optimized, Inhalable, Gemcitabine-Loaded Gelatin Nanocarriers for Lung Cancer. J. Aerosol Med. Pulm. Drug Deliv. 2017;30:299–321. doi: 10.1089/jamp.2015.1286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 312.Kozak M. An analysis of 5′-noncoding sequences from 699 vertebrate messenger RNAs. Nucleic Acids Res. 1987;15:8125–8148. doi: 10.1093/nar/15.20.8125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 313.Lai E.C., Posakony J.W. The Bearded box, a novel 3′ UTR sequence motif, mediates negative post-transcriptional regulation of Bearded and Enhancer of split Complex gene expression. Development. 1997;124:4847–4856. doi: 10.1242/dev.124.23.4847. [DOI] [PubMed] [Google Scholar]
  • 314.Desrosiers R., Friderici K., Rottman F. Identification of methylated nucleosides in messenger RNA from Novikoff hepatoma cells. Proc. Natl. Acad. Sci. USA. 1974;71:3971–3975. doi: 10.1073/pnas.71.10.3971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 315.Patel A.K., Kaczmarek J.C., Bose S., Kauffman K.J., Mir F., Heartlein M.W., DeRosa F., Langer R., Anderson D.G. Inhaled Nanoformulated mRNA Polyplexes for Protein Production in Lung Epithelium. Adv. Mater. 2019;31:e1805116. doi: 10.1002/adma.201805116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 316.Li J., Wang W., He Y., Li Y., Yan E.Z., Zhang K., Irvine D.J., Hammond P.T. Structurally Programmed Assembly of Translation Initiation Nanoplex for Superior mRNA Delivery. ACS Nano. 2017;11:2531–2544. doi: 10.1021/acsnano.6b08447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 317.Wang X., Lu Z., Gomez A., Hon G.C., Yue Y., Han D., Fu Y., Parisien M., Dai Q., Jia G. N6-methyladenosine-dependent regulation of messenger RNA stability. Nature. 2014;505:117–120. doi: 10.1038/nature12730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 318.Agu R.U., Ugwoke M.I., Armand M., Kinget R., Verbeke N. The lung as a route for systemic delivery of therapeutic proteins and peptides. Respir. Res. 2001;2:198–209. doi: 10.1186/rr58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 319.Kreyling W.G., Hirn S., Schleh C. Nanoparticles in the lung. Nat. Biotechnol. 2010;28:1275–1276. doi: 10.1038/nbt.1735. [DOI] [PubMed] [Google Scholar]
  • 320.Sijts A.J.A.M., Standera S., Toes R.E.M., Ruppert T., Beekman N.J.C.M., van Veelen P.A., Ossendorp F.A., Melief C.J., Kloetzel P.M. MHC class I antigen processing of an adenovirus CTL epitope is linked to the levels of immunoproteasomes in infected cells. J. Immunol. 2000;164:4500–4506. doi: 10.4049/jimmunol.164.9.4500. [DOI] [PubMed] [Google Scholar]
  • 321.Kaczmarek J.C., Kowalski P.S., Anderson D.G. Advances in the delivery of RNA therapeutics: from concept to clinical reality. Genome Med. 2017;9:60. doi: 10.1186/s13073-017-0450-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 322.Muthukrishnan S., Both G.W., Furuichi Y., Shatkin A.J. 5′-Terminal 7-methylguanosine in eukaryotic mRNA is required for translation. Nature. 1975;255:33–37. doi: 10.1038/255033a0. [DOI] [PubMed] [Google Scholar]
  • 323.Vachon V.K., Calderon B.M., Conn G.L. A novel RNA molecular signature for activation of 2′-5′ oligoadenylate synthetase-1. Nucleic Acids Res. 2015;43:544–552. doi: 10.1093/nar/gku1289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 324.Luthra P., Sun D., Silverman R.H., He B. Activation of IFN-β expression by a viral mRNA through RNase L and MDA5. Proc. Natl. Acad. Sci. USA. 2011;108:2118–2123. doi: 10.1073/pnas.1012409108. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Molecular Therapy are provided here courtesy of The American Society of Gene & Cell Therapy

RESOURCES