Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(11)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38892347

ABSTRACT

V-set immunoglobulin domain-containing 4 (VSIG4) is a B7 family protein with known roles as a C3 fragment complement receptor involved in pathogen clearance and a negative regulator of T cell activation by an undetermined mechanism. VSIG4 expression is specific for tumor-associated and select tissue-resident macrophages. Increased expression of VSIG4 has been associated with worse survival in multiple cancer indications. Based upon computational analysis of transcript data across thousands of tumor and normal tissue samples, we hypothesized that VSIG4 has an important role in promoting M2-like immune suppressive macrophages and that targeting VSIG4 could relieve VSIG4-mediated macrophage suppression by repolarizing tumor-associated macrophages (TAMs) to an inflammatory phenotype. We have also observed a cancer-specific pattern of VSIG4 isoform distribution, implying a change in the functional regulation in cancer. Through a series of in vitro, in vivo, and ex vivo assays we demonstrate that anti-VSIG4 antibodies repolarize M2 macrophages and induce an immune response culminating in T cell activation. Anti-VSIG4 antibodies induce pro-inflammatory cytokines in M-CSF plus IL-10-driven human monocyte-derived M2c macrophages. Across patient-derived tumor samples from multiple tumor types, anti-VSIG4 treatment resulted in the upregulation of cytokines associated with TAM repolarization and T cell activation and chemokines involved in immune cell recruitment. VSIG4 blockade is also efficacious in a syngeneic mouse model as monotherapy as it enhances efficacy in combination with anti-PD-1, and the effect is dependent on the systemic availability of CD8+ T cells. Thus, VSIG4 represents a promising new target capable of triggering an anti-cancer response via multiple key immune mechanisms.


Subject(s)
Neoplasms , Tumor-Associated Macrophages , Animals , Humans , Mice , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Cell Line, Tumor , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Cytokines/metabolism , Female , Receptors, Complement
2.
Cell Rep ; 42(5): 112372, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37086404

ABSTRACT

Autophagy is a homeostatic process critical for cellular survival, and its malfunction is implicated in human diseases including neurodegeneration. Loss of autophagy contributes to cytotoxicity and tissue degeneration, but the mechanistic understanding of this phenomenon remains elusive. Here, we generated autophagy-deficient (ATG5-/-) human embryonic stem cells (hESCs), from which we established a human neuronal platform to investigate how loss of autophagy affects neuronal survival. ATG5-/- neurons exhibit basal cytotoxicity accompanied by metabolic defects. Depletion of nicotinamide adenine dinucleotide (NAD) due to hyperactivation of NAD-consuming enzymes is found to trigger cell death via mitochondrial depolarization in ATG5-/- neurons. Boosting intracellular NAD levels improves cell viability by restoring mitochondrial bioenergetics and proteostasis in ATG5-/- neurons. Our findings elucidate a mechanistic link between autophagy deficiency and neuronal cell death that can be targeted for therapeutic interventions in neurodegenerative and lysosomal storage diseases associated with autophagic defect.


Subject(s)
NAD , Nicotinamide Mononucleotide , Humans , NAD/metabolism , Nicotinamide Mononucleotide/metabolism , Neurons/metabolism , Mitochondria/metabolism , Autophagy , Niacinamide/metabolism
3.
Mol Ther Nucleic Acids ; 19: 252-266, 2020 Mar 06.
Article in English | MEDLINE | ID: mdl-31855834

ABSTRACT

Translation is an essential biological process, and dysregulation is associated with a range of diseases including ribosomopathies, diabetes, and cancer. Here, we examine translation dysregulation in vivo using RNAi to knock down the m-subunit of the translation initiation factor eIF3 in the mouse liver. Transcriptome sequencing, ribosome profiling, whole proteome, and phosphoproteome analyses show that eIF3m deficiency leads to the transcriptional response and changes in cellular translation that yield few detectable differences in the translation of particular mRNAs. The transcriptional response fell into two main categories: ribosome biogenesis (increased transcription of ribosomal proteins) and cell metabolism (alterations in lipid, amino acid, nucleic acid, and drug metabolism). Analysis of ribosome biogenesis reveals inhibition of rRNA processing, highlighting decoupling of rRNA synthesis and ribosomal protein gene transcription in response to eIF3m knockdown. Interestingly, a similar reduction in eIF3m protein levels is associated with induction of the mTOR pathway in vitro but not in vivo. Overall, this work highlights the utility of a RNAi-based in vivo approach for studying the regulation of mammalian translation in vivo.

4.
Nat Metab ; 1(9): 912-926, 2019 09.
Article in English | MEDLINE | ID: mdl-31572976

ABSTRACT

Atherosclerosis is a progressive vascular disease triggered by interplay between abnormal shear stress and endothelial lipid retention. A combination of these and, potentially, other factors leads to a chronic inflammatory response in the vessel wall, which is thought to be responsible for disease progression characterized by a buildup of atherosclerotic plaques. Yet molecular events responsible for maintenance of plaque inflammation and plaque growth have not been fully defined. Here we show that endothelial TGFß signaling is one of the primary drivers of atherosclerosis-associated vascular inflammation. Inhibition of endothelial TGFß signaling in hyperlipidemic mice reduces vessel wall inflammation and vascular permeability and leads to arrest of disease progression and regression of established lesions. These pro-inflammatory effects of endothelial TGFß signaling are in stark contrast with its effects in other cell types and identify it as an important driver of atherosclerotic plaque growth and show the potential of cell-type specific therapeutic intervention aimed at control of this disease.


Subject(s)
Atherosclerosis/metabolism , Endothelium, Vascular/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Vasculitis/metabolism , Animals , Capillary Permeability , Cell Line , Disease Progression , Endothelium, Vascular/pathology , Humans , Mice , Mice, Knockout , Transforming Growth Factor beta/genetics
5.
Adv Mater ; 31(8): e1805116, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30609147

ABSTRACT

Noninvasive aerosol inhalation is an established method of drug delivery to the lung, and remains a desirable route for nucleic-acid-based therapeutics. In vitro transcribed (IVT) mRNA has broad therapeutic applicability as it permits temporal and dose-dependent control of encoded protein expression. Inhaled delivery of IVT-mRNA has not yet been demonstrated and requires development of safe and effective materials. To meet this need, hyperbranched poly(beta amino esters) (hPBAEs) are synthesized to enable nanoformulation of stable and concentrated polyplexes suitable for inhalation. This strategy achieves uniform distribution of luciferase mRNA throughout all five lobes of the lung and produces 101.2 ng g-1 of luciferase protein 24 h after inhalation of hPBAE polyplexes. Importantly, delivery is localized to the lung, and no luminescence is observed in other tissues. Furthermore, using an Ai14 reporter mouse model it is identified that 24.6% of the total lung epithelial cell population is transfected after a single dose. Repeat dosing of inhaled hPBAE-mRNA generates consistent protein production in the lung, without local or systemic toxicity. The results indicate that nebulized delivery of IVT-mRNA facilitated by hPBAE vectors may provide a clinically relevant delivery system to lung epithelium.


Subject(s)
Epithelial Cells/metabolism , Luciferases/genetics , Nanoparticles/chemistry , Polymers/chemistry , RNA, Messenger/chemistry , Administration, Inhalation , Animals , Drug Compounding/methods , Drug Liberation , Epithelial Cells/cytology , Female , Gene Transfer Techniques , Genetic Therapy/methods , Hydrogen-Ion Concentration , Lung/drug effects , Mice , Mice, Inbred C57BL , Models, Animal , RNA, Messenger/administration & dosage , RNA, Messenger/adverse effects , RNA, Messenger/metabolism , Tissue Distribution , Transfection/methods
6.
Circulation ; 139(1): 78-96, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30586693

ABSTRACT

BACKGROUND: Chronic kidney disease (CKD) increases cardiovascular risk. Underlying mechanisms, however, remain obscure. The uremic toxin indoxyl sulfate is an independent cardiovascular risk factor in CKD. We explored the potential impact of indoxyl sulfate on proinflammatory activation of macrophages and its underlying mechanisms. METHODS: We examined in vitro the effects of clinically relevant concentrations of indoxyl sulfate on proinflammatory responses of macrophages and the roles of organic anion transporters and organic anion transporting polypeptides (OATPs). A systems approach, involving unbiased global proteomics, bioinformatics, and network analysis, then explored potential key pathways. To address the role of Delta-like 4 (Dll4) in indoxyl sulfate-induced macrophage activation and atherogenesis in CKD in vivo, we used 5/6 nephrectomy and Dll4 antibody in low-density lipoprotein receptor-deficient (Ldlr-/-) mice. To further determine the relative contribution of OATP2B1 or Dll4 to proinflammatory activation of macrophages and atherogenesis in vivo, we used siRNA delivered by macrophage-targeted lipid nanoparticles in mice. RESULTS: We found that indoxyl sulfate-induced proinflammatory macrophage activation is mediated by its uptake through transporters, including OATP2B1, encoded by the SLCO2B1 gene. The global proteomics identified potential mechanisms, including Notch signaling and the ubiquitin-proteasome pathway, that mediate indoxyl sulfate-triggered proinflammatory macrophage activation. We chose the Notch pathway as an example of key candidates for validation of our target discovery platform and for further mechanistic studies. As predicted computationally, indoxyl sulfate triggered Notch signaling, which was preceded by the rapid induction of Dll4 protein. Dll4 induction may result from inhibition of the ubiquitin-proteasome pathway, via the deubiquitinating enzyme USP5. In mice, macrophage-targeted OATP2B1/Slco2b1 silencing and Dll4 antibody inhibited proinflammatory activation of peritoneal macrophages induced by indoxyl sulfate. In low-density lipoprotein receptor-deficient mice, Dll4 antibody abolished atherosclerotic lesion development accelerated in Ldlr-/- mice. Moreover, coadministration of indoxyl sulfate and OATP2B1/Slco2b1 or Dll4 siRNA encapsulated in macrophage-targeted lipid nanoparticles in Ldlr-/- mice suppressed lesion development. CONCLUSIONS: These results suggest that novel crosstalk between OATP2B1 and Dll4-Notch signaling in macrophages mediates indoxyl sulfate-induced vascular inflammation in CKD.


Subject(s)
Atherosclerosis/metabolism , Indican/toxicity , Intercellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Macrophage Activation/drug effects , Macrophages/drug effects , Membrane Proteins/metabolism , Organic Anion Transporters/metabolism , Receptors, Notch/metabolism , Renal Insufficiency, Chronic/metabolism , Adaptor Proteins, Signal Transducing , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Calcium-Binding Proteins , Disease Models, Animal , Humans , Inflammation Mediators/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Macrophages/metabolism , Macrophages/pathology , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , Organic Anion Transporters/genetics , Phenotype , Plaque, Atherosclerotic , RAW 264.7 Cells , Receptors, LDL/deficiency , Receptors, LDL/genetics , Receptors, Notch/genetics , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Signal Transduction/drug effects , Vascular Calcification/metabolism , Vascular Calcification/pathology
7.
Nano Lett ; 18(10): 6449-6454, 2018 10 10.
Article in English | MEDLINE | ID: mdl-30211557

ABSTRACT

mRNA therapeutics hold great potential for treating a variety of diseases through protein-replacement, immunomodulation, and gene editing. However, much like siRNA therapy the majority of progress in mRNA delivery has been confined to the liver. Previously, we demonstrated that poly(ß-amino esters), a class of degradable polymers, are capable of systemic mRNA delivery to the lungs in mice when formulated into nanoparticles with poly(ethylene glycol)-lipid conjugates. Using experimental design, a statistical approach to optimization that reduces experimental burden, we demonstrate herein that these degradable polymer-lipid nanoparticles can be optimized in terms of polymer synthesis and nanoparticle formulation to achieve a multiple order-of-magnitude increase in potency. Furthermore, using genetically engineered Cre reporter mice, we demonstrate that mRNA is functionally delivered to both the lung endothelium and pulmonary immune cells, expanding the potential utility of these nanoparticles.


Subject(s)
Endothelium/drug effects , Lung/drug effects , Nanoparticles/administration & dosage , RNA, Small Interfering/administration & dosage , Animals , Endothelium/immunology , Endothelium/pathology , Gene Transfer Techniques , Humans , Lipids/administration & dosage , Lipids/chemistry , Lung/immunology , Lung/pathology , Mice , Nanoparticles/chemistry , Polyethylene Glycols/chemistry , Polymers/chemistry , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics
8.
Angew Chem Int Ed Engl ; 57(41): 13582-13586, 2018 10 08.
Article in English | MEDLINE | ID: mdl-30112821

ABSTRACT

RNAs are a promising class of therapeutics given their ability to regulate protein concentrations at the cellular level. Developing safe and effective strategies to deliver RNAs remains important for realizing their full clinical potential. Here, we develop lipid nanoparticle formulations that can deliver short interfering RNAs (for gene silencing) or messenger RNAs (for gene upregulation). Specifically, we study how the tail length, tail geometry, and linker spacing in diketopiperazine lipid materials influences LNP potency with siRNAs and mRNAs. Eight lipid materials are synthesized, and 16 total formulations are screened for activity in vitro; the lead material is evaluated with mRNA for in vivo use and demonstrates luciferase protein expression in the spleen. In undertaking this approach, not only do we develop synthetic routes to delivery materials, but we also reveal structural criteria that could be useful for developing next-generation delivery materials for RNA therapeutics.


Subject(s)
Lipids/chemistry , Nanoparticles/chemistry , RNA, Messenger/administration & dosage , RNA, Small Interfering/administration & dosage
9.
Mol Ther Nucleic Acids ; 10: 55-63, 2018 Mar 02.
Article in English | MEDLINE | ID: mdl-29499956

ABSTRACT

mRNA therapeutics hold promise for the treatment of diseases requiring intracellular protein expression and for use in genome editing systems, but mRNA must transfect the desired tissue and cell type to be efficacious. Nanoparticle vectors that deliver the mRNA are often evaluated using mRNA encoding for reporter genes such as firefly luciferase (FLuc); however, single-cell resolution of mRNA expression cannot generally be achieved with FLuc, and, thus, the transfected cell populations cannot be determined without additional steps or experiments. To more rapidly identify which types of cells an mRNA formulation transfects in vivo, we describe a Cre recombinase (Cre)-based system that permanently expresses fluorescent tdTomato protein in transfected cells of genetically modified mice. Following in vivo application of vectored Cre mRNA, it is possible to visualize successfully transfected cells via Cre-mediated tdTomato expression in bulk tissues and with single-cell resolution. Using this system, we identify previously unknown transfected cell types of an existing mRNA delivery vehicle in vivo and also develop a new mRNA formulation capable of transfecting lung endothelial cells. Importantly, the same formulations with mRNA encoding for fluorescent protein delivered to wild-type mice did not produce sufficient signal for any visualization in vivo, demonstrating the significantly improved sensitivity of our Cre-based system. We believe that the system described here may facilitate the identification and characterization of mRNA delivery vectors to new tissues and cell types.

10.
Adv Mater ; 29(33)2017 Sep.
Article in English | MEDLINE | ID: mdl-28681930

ABSTRACT

B lymphocytes regulate several aspects of immunity including antibody production, cytokine secretion, and T-cell activation; moreover, B cell misregulation is implicated in autoimmune disorders and cancers such as multiple sclerosis and non-Hodgkin's lymphomas. The delivery of messenger RNA (mRNA) into B cells can be used to modulate and study these biological functions by means of inducing functional protein expression in a dose-dependent and time-controlled manner. However, current in vivo mRNA delivery systems fail to transfect B lymphocytes and instead primarily target hepatocytes and dendritic cells. Here, the design, synthesis, and biological evaluation of a lipid nanoparticle (LNP) system that can encapsulate mRNA, navigate to the spleen, transfect B lymphocytes, and induce more than 60 pg of protein expression per million B cells within the spleen is described. Importantly, this LNP induces more than 85% of total protein production in the spleen, despite LNPs being observed transiently in the liver and other organs. These results demonstrate that LNP composition alone can be used to modulate the site of protein induction in vivo, highlighting the critical importance of designing and synthesizing new nanomaterials for nucleic acid delivery.


Subject(s)
Lipids/chemistry , B-Lymphocytes , Liver , Nanoparticles , RNA, Messenger
11.
Mol Ther Nucleic Acids ; 7: 314-323, 2017 Jun 16.
Article in English | MEDLINE | ID: mdl-28624207

ABSTRACT

Fibrotic diseases contribute to 45% of deaths in the industrialized world, and therefore a better understanding of the pathophysiological mechanisms underlying tissue fibrosis is sorely needed. We aimed to identify novel modifiers of tissue fibrosis expressed by myofibroblasts and their progenitors in their disease microenvironment through RNA silencing in vivo. We leveraged novel biology, targeting genes upregulated during liver and kidney fibrosis in this cell lineage, and employed small interfering RNA (siRNA)-formulated lipid nanoparticles technology to silence these genes in carbon-tetrachloride-induced liver fibrosis in mice. We identified five genes, Egr2, Atp1a2, Fkbp10, Fstl1, and Has2, which modified fibrogenesis based on their silencing, resulting in reduced Col1a1 mRNA levels and collagen accumulation in the liver. These genes fell into different groups based on the effects of their silencing on a transcriptional mini-array and histological outcomes. Silencing of Egr2 had the broadest effects in vivo and also reduced fibrogenic gene expression in a human fibroblast cell line. Prior to our study, Egr2, Atp1a2, and Fkbp10 had not been functionally validated in fibrosis in vivo. Thus, our results provide a major advance over the existing knowledge of fibrogenic pathways. Our study is the first example of a targeted siRNA assay to identify novel fibrosis modifiers in vivo.

12.
Cancer Cell ; 31(4): 576-590.e8, 2017 04 10.
Article in English | MEDLINE | ID: mdl-28399412

ABSTRACT

Cyclins and cyclin-dependent kinases (CDKs) are hyperactivated in numerous human tumors. To identify means of interfering with cyclins/CDKs, we performed nine genome-wide screens for human microRNAs (miRNAs) directly regulating cell-cycle proteins. We uncovered a distinct class of miRNAs that target nearly all cyclins/CDKs, which are very effective in inhibiting cancer cell proliferation. By profiling the response of over 120 human cancer cell lines, we derived an expression-based algorithm that can predict the response of tumors to cell-cycle-targeting miRNAs. Using systemic administration of nanoparticle-formulated miRNAs, we inhibited tumor progression in seven mouse xenograft models, including three treatment-refractory patient-derived tumors, without affecting normal tissues. Our results highlight the utility of using cell-cycle-targeting miRNAs for treatment of refractory cancer types.


Subject(s)
Cell Cycle/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , 3' Untranslated Regions , Algorithms , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Drug Delivery Systems/methods , Female , Genome-Wide Association Study , Humans , Mice, Inbred Strains , MicroRNAs/administration & dosage , MicroRNAs/pharmacology , Mutation , Nanoparticles , Proto-Oncogene Proteins p21(ras)/genetics , Xenograft Model Antitumor Assays
13.
Nat Rev Drug Discov ; 16(6): 387-399, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28337020

ABSTRACT

With the recent development of CRISPR technology, it is becoming increasingly easy to engineer the genome. Genome-editing systems based on CRISPR, as well as transcription activator-like effector nucleases (TALENs) and zinc-finger nucleases (ZFNs), are becoming valuable tools for biomedical research, drug discovery and development, and even gene therapy. However, for each of these systems to effectively enter cells of interest and perform their function, efficient and safe delivery technologies are needed. This Review discusses the principles of biomacromolecule delivery and gene editing, examines recent advances and challenges in non-viral and viral delivery methods, and highlights the status of related clinical trials.


Subject(s)
Gene Editing/methods , Genetic Engineering/methods , Genetic Therapy/methods , Genome, Human/genetics , Animals , Biomedical Research/methods , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Drug Discovery/methods , Gene Transfer Techniques , Humans , Transcription Activator-Like Effector Nucleases/genetics , Zinc Fingers/genetics
14.
Proc Natl Acad Sci U S A ; 114(8): 2060-2065, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28167778

ABSTRACT

Nucleic acid therapeutics are limited by inefficient delivery to target tissues and cells and by an incomplete understanding of how nanoparticle structure affects biodistribution to off-target organs. Although thousands of nanoparticle formulations have been designed to deliver nucleic acids, most nanoparticles have been tested in cell culture contexts that do not recapitulate systemic in vivo delivery. To increase the number of nanoparticles that could be tested in vivo, we developed a method to simultaneously measure the biodistribution of many chemically distinct nanoparticles. We formulated nanoparticles to carry specific nucleic acid barcodes, administered the pool of particles, and quantified particle biodistribution by deep sequencing the barcodes. This method distinguished previously characterized lung- and liver- targeting nanoparticles and accurately reported relative quantities of nucleic acid delivered to tissues. Barcode sequences did not affect delivery, and no evidence of particle mixing was observed for tested particles. By measuring the biodistribution of 30 nanoparticles to eight tissues simultaneously, we identified chemical properties promoting delivery to some tissues relative to others. Finally, particles that distributed to the liver also silenced gene expression in hepatocytes when formulated with siRNA. This system can facilitate discovery of nanoparticles targeting specific tissues and cells and accelerate the study of relationships between chemical structure and delivery in vivo.


Subject(s)
DNA Barcoding, Taxonomic/methods , Drug Discovery/methods , Nanoparticles/chemistry , Nucleic Acids/administration & dosage , RNA, Small Interfering/administration & dosage , Animals , Cell Separation , Drug Delivery Systems/methods , Factor VII/genetics , Female , Flow Cytometry , Liver/cytology , Liver/drug effects , Lung/cytology , Lung/drug effects , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy/methods , Nucleic Acids/therapeutic use , Pharmaceutical Preparations/administration & dosage , RNA Interference , RNA, Small Interfering/therapeutic use , Tissue Distribution
15.
Angew Chem Int Ed Engl ; 55(44): 13808-13812, 2016 10 24.
Article in English | MEDLINE | ID: mdl-27690187

ABSTRACT

Therapeutic nucleic acids hold great promise for the treatment of disease but require vectors for safe and effective delivery. Synthetic nanoparticle vectors composed of poly(ß-amino esters) (PBAEs) and nucleic acids have previously demonstrated potential utility for local delivery applications. To expand this potential utility to include systemic delivery of mRNA, hybrid polymer-lipid nanoformulations for systemic delivery to the lungs were developed. Through coformulation of PBAEs with lipid-polyethylene glycol (PEG), mRNA formulations were developed with increased serum stability and increased in vitro potency. The formulations were capable of functional delivery of mRNA to the lungs after intravenous administration in mice. To our knowledge, this is the first report of the systemic administration of mRNA for delivery to the lungs using degradable polymer-lipid nanoparticles.


Subject(s)
Lipids/chemistry , Lung/chemistry , Nanoparticles/chemistry , Polymers/chemistry , RNA, Messenger/chemistry , Administration, Intravenous , Animals , Mice , Molecular Structure , Particle Size , Polymers/administration & dosage , RNA, Messenger/administration & dosage , RNA, Messenger/chemical synthesis , Surface Properties
16.
Biomaterials ; 109: 78-87, 2016 12.
Article in English | MEDLINE | ID: mdl-27680591

ABSTRACT

mRNA has broad potential for treating diseases requiring protein expression. However, mRNA can also induce an immune response with associated toxicity. Replacement of uridine bases with pseudouridine has been postulated to modulate both mRNA immunogenicity and potency. Here, we explore the immune response and activity of lipid nanoparticle-formulated unmodified and pseudouridine-modified mRNAs administered systemically in vivo. Pseudouridine modification to mRNA had no significant effect on lipid nanoparticle physical properties, protein expression in vivo, or mRNA immunogenicity compared to unmodified mRNA when delivered systemically with liver-targeting lipid nanoparticles, but reduced in vitro transfection levels. Indicators of a transient, extracellular innate immune response to mRNA were observed, including neutrophilia, myeloid cell activation, and up-regulation of four serum cytokines. This study provides insight into the immune responses to mRNA lipid nanoparticles, and suggests that pseudouridine modifications may be unnecessary for therapeutic application of mRNA in the liver.


Subject(s)
Lipids/chemistry , Nanoparticles/chemistry , Pseudouridine/chemistry , RNA, Messenger/chemistry , Animals , Cytokines/metabolism , Female , Gene Expression , Gene Transfer Techniques , HeLa Cells , Humans , Immunity, Innate , Liver/metabolism , Mice, Inbred C57BL , Myeloid Cells/immunology , Nanoparticles/administration & dosage , Particle Size , RNA, Messenger/administration & dosage , RNA, Messenger/biosynthesis , RNA, Messenger/immunology , Surface Properties , Transfection
17.
Sci Transl Med ; 8(342): 342ra80, 2016 06 08.
Article in English | MEDLINE | ID: mdl-27280687

ABSTRACT

Myocardial infarction (MI) leads to a systemic surge of vascular inflammation in mice and humans, resulting in secondary ischemic complications and high mortality. We show that, in ApoE(-/-) mice with coronary ligation, increased sympathetic tone up-regulates not only hematopoietic leukocyte production but also plaque endothelial expression of adhesion molecules. To counteract the resulting arterial leukocyte recruitment, we developed nanoparticle-based RNA interference (RNAi) that effectively silences five key adhesion molecules. Simultaneously encapsulating small interfering RNA (siRNA)-targeting intercellular cell adhesion molecules 1 and 2 (Icam1 and Icam2), vascular cell adhesion molecule 1 (Vcam1), and E- and P-selectins (Sele and Selp) into polymeric endothelial-avid nanoparticles reduced post-MI neutrophil and monocyte recruitment into atherosclerotic lesions and decreased matrix-degrading plaque protease activity. Five-gene combination RNAi also curtailed leukocyte recruitment to ischemic myocardium. Therefore, targeted multigene silencing may prevent complications after acute MI.


Subject(s)
Cell Adhesion Molecules/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/therapy , Neutrophil Infiltration/physiology , RNA, Small Interfering/genetics , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cell Adhesion Molecules/genetics , E-Selectin/genetics , E-Selectin/metabolism , Female , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , Myocardial Infarction/immunology , Nanoparticles , Neutrophil Infiltration/genetics , P-Selectin/genetics , P-Selectin/metabolism , Parabiosis , RNA Interference , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
18.
Adv Mater ; 28(15): 2939-43, 2016 Apr 20.
Article in English | MEDLINE | ID: mdl-26889757

ABSTRACT

Thousands of human diseases could be treated by selectively controlling the expression of specific proteins in vivo. A new series of alkenyl amino alcohol (AAA) ionizable lipid nanoparticles (LNPs) capable of delivering human mRNA with unprecedented levels of in vivo efficacy is demonstrated. This study highlights the importance of utilizing synthesis tools in tandem with biological inspiration to understand and improve nucleic acid delivery in vivo.


Subject(s)
Alkenes/chemistry , Amino Alcohols/chemistry , Biomimetic Materials/chemistry , Drug Carriers/chemistry , Lipids/chemistry , Nanoparticles/chemistry , Erythropoietin/genetics , Humans , RNA, Messenger/chemistry , RNA, Messenger/genetics
19.
J Control Release ; 240: 227-234, 2016 10 28.
Article in English | MEDLINE | ID: mdl-26718856

ABSTRACT

Though therapeutics based on messenger RNA (mRNA) have broad potential in applications such as protein replacement therapy, cancer immunotherapy, and genomic engineering, their effective intracellular delivery remains a challenge. A chemically diverse suite of delivery materials with origins as materials for cellular transfection of DNA and small interfering RNAs (siRNAs) has recently been reported to have promise as non-viral delivery agents for mRNA. These materials include covalent conjugates, protamine complexes, nanoparticles based on lipids or polymers, and hybrid formulations. This review will highlight the use of delivery materials for mRNA, with a specific focus on their mechanisms of action, routes of administration, and dosages. Additionally, strategies in which these materials can be adapted and optimized to address challenges specific to mRNA delivery are also discussed. The technologies included have shown varying promise for therapeutic use, specifically having been used to deliver mRNA in vivo or exhibiting characteristics that could make in vivo use a possibility. In so doing, it is the intention of this review to provide a comprehensive look at the progress and possibilities in applying nucleic acid delivery technology specifically toward the emerging area of mRNA therapeutics.


Subject(s)
Drug Delivery Systems/methods , Genetic Therapy/methods , Intracellular Fluid/drug effects , RNA, Messenger/administration & dosage , Animals , Drug Delivery Systems/trends , Genetic Therapy/trends , Humans , Intracellular Fluid/metabolism , Polymers/administration & dosage , Polymers/metabolism , Protamines/administration & dosage , Protamines/genetics , Protamines/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
20.
Nano Lett ; 15(11): 7300-6, 2015 Nov 11.
Article in English | MEDLINE | ID: mdl-26469188

ABSTRACT

Intracellular delivery of messenger RNA (mRNA) has the potential to induce protein production for many therapeutic applications. Although lipid nanoparticles have shown considerable promise for the delivery of small interfering RNAs (siRNA), their utility as agents for mRNA delivery has only recently been investigated. The most common siRNA formulations contain four components: an amine-containing lipid or lipid-like material, phospholipid, cholesterol, and lipid-anchored polyethylene glycol, the relative ratios of which can have profound effects on the formulation potency. Here, we develop a generalized strategy to optimize lipid nanoparticle formulations for mRNA delivery to the liver in vivo using Design of Experiment (DOE) methodologies including Definitive Screening and Fractional Factorial Designs. By simultaneously varying lipid ratios and structures, we developed an optimized formulation which increased the potency of erythropoietin-mRNA-loaded C12-200 lipid nanoparticles 7-fold relative to formulations previously used for siRNA delivery. Key features of this optimized formulation were the incorporation of 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) and increased ionizable lipid:mRNA weight ratios. Interestingly, the optimized lipid nanoparticle formulation did not improve siRNA delivery, indicating differences in optimized formulation parameter design spaces for siRNA and mRNA. We believe the general method described here can accelerate in vivo screening and optimization of nanoparticle formulations with large multidimensional design spaces.


Subject(s)
Gene Transfer Techniques , Lipids/chemistry , Nanoparticles/chemistry , RNA, Messenger/administration & dosage , Cell Line, Tumor , Humans , Lipids/administration & dosage , Liposomes/administration & dosage , Liposomes/chemistry , Liver/drug effects , Nanoparticles/administration & dosage , Phosphatidylethanolamines/administration & dosage , Phosphatidylethanolamines/chemistry , Polyethylene Glycols/chemistry , RNA, Messenger/chemistry , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...