Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Cell ; 184(13): 3410-3425.e17, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34062120

ABSTRACT

To control viral infection, vertebrates rely on both inducible interferon responses and less well-characterized cell-intrinsic responses composed of "at the ready" antiviral effector proteins. Here, we show that E3 ubiquitin ligase TRIM7 is a cell-intrinsic antiviral effector that restricts multiple human enteroviruses by targeting viral 2BC, a membrane remodeling protein, for ubiquitination and proteasome-dependent degradation. Selective pressure exerted by TRIM7 results in emergence of a TRIM7-resistant coxsackievirus with a single point mutation in the viral 2C ATPase/helicase. In cultured cells, the mutation helps the virus evade TRIM7 but impairs optimal viral replication, and this correlates with a hyperactive and structurally plastic 2C ATPase. Unexpectedly, the TRIM7-resistant virus has a replication advantage in mice and causes lethal pancreatitis. These findings reveal a unique mechanism for targeting enterovirus replication and provide molecular insight into the benefits and trade-offs of viral evolution imposed by a host restriction factor.


Subject(s)
Enterovirus/physiology , Enterovirus/pathogenicity , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Virus Replication/physiology , Adenosine Triphosphatases/metabolism , Animals , Cell Line , Female , Humans , Inflammation/pathology , Mice, Inbred C57BL , Mutation/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Proteolysis , RNA, Viral/metabolism , Ubiquitin/metabolism , Viral Proteins/genetics
2.
Proc Natl Acad Sci U S A ; 120(52): e2313009120, 2023 Dec 26.
Article in English | MEDLINE | ID: mdl-38109533

ABSTRACT

Genetic medicines have the potential to treat various diseases; however, certain ailments including inflammatory diseases and cancer would benefit from control over extracellular localization of therapeutic proteins. A critical gap therefore remains the need to develop and incorporate methodologies that allow for posttranslational control over expression dynamics, localization, and stability of nucleic acid-generated protein therapeutics. To address this, we explored how the body's endogenous machinery controls protein localization through signal peptides (SPs), including how these motifs could be incorporated modularly into therapeutics. SPs serve as a virtual zip code for mRNA transcripts that direct the cell where to send completed proteins within the cell and the body. Utilizing this signaling biology, we incorporated secretory SP sequences upstream of mRNA transcripts coding for reporter, natural, and therapeutic proteins to induce secretion of the proteins into systemic circulation. SP sequences generated secretion of various engineered proteins into the bloodstream following intravenous, intramuscular, and subcutaneous SP mRNA delivery by lipid, polymer, and ionizable phospholipid delivery carriers. SP-engineered etanercept/TNF-α inhibitor proteins demonstrated therapeutic efficacy in an imiquimod-induced psoriasis model by reducing hyperkeratosis and inflammation. An SP-engineered anti-PD-L1 construct mediated mRNA encoded proteins with longer serum half-lives that reduced tumor burden and extended survival in MC38 and B16F10 cancer models. The modular nature of SP platform should enable intracellular and extracellular localization control of various functional proteins for diverse therapeutic applications.


Subject(s)
Dermatitis , Melanoma , Psoriasis , Humans , Animals , Melanoma/drug therapy , Melanoma/genetics , Psoriasis/drug therapy , Psoriasis/genetics , Inflammation/pathology , Protein Sorting Signals , RNA, Messenger/genetics , Disease Models, Animal
3.
Hepatology ; 78(4): 1133-1148, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37039560

ABSTRACT

BACKGROUND AND AIMS: The liver is remarkably regenerative and can completely recover even when 80% of its mass is surgically removed. Identification of secreted factors that regulate liver growth would help us understand how organ size and regeneration are controlled but also provide candidate targets to promote regeneration or impair cancer growth. APPROACH AND RESULTS: To enrich for secreted factors that regulate growth control, we induced massive liver overgrowth with either YAP or MYC . Differentially expressed secreted factors were identified in these livers using transcriptomic analysis. To rank candidates by functionality, we performed in vivo CRISPR screening using the Fah knockout model of tyrosinemia. We identified secreted phosphoprotein-2 (SPP2) as a secreted factor that negatively regulates regeneration. Spp2 -deficient mice showed increased survival after acetaminophen poisoning and reduced fibrosis after repeated carbon tetrachloride injections. We examined the impact of SPP2 on bone morphogenetic protein signaling in liver cells and found that SPP2 antagonized bone morphogenetic protein signaling in vitro and in vivo. We also identified cell-surface receptors that interact with SPP2 using a proximity biotinylation assay coupled with mass spectrometry. We showed that SPP2's interactions with integrin family members are in part responsible for some of the regeneration phenotypes. CONCLUSIONS: Using an in vivo CRISPR screening system, we identified SPP2 as a secreted factor that negatively regulates liver regeneration. This study provides ways to identify, validate, and characterize secreted factors in vivo.


Subject(s)
Liver Regeneration , Neoplasms , Mice , Animals , Liver/metabolism , Hepatocytes/metabolism , Signal Transduction
4.
Proc Natl Acad Sci U S A ; 118(52)2021 12 28.
Article in English | MEDLINE | ID: mdl-34933999

ABSTRACT

Lipid nanoparticles (LNPs) are a clinically mature technology for the delivery of genetic medicines but have limited therapeutic applications due to liver accumulation. Recently, our laboratory developed selective organ targeting (SORT) nanoparticles that expand the therapeutic applications of genetic medicines by enabling delivery of messenger RNA (mRNA) and gene editing systems to non-liver tissues. SORT nanoparticles include a supplemental SORT molecule whose chemical structure determines the LNP's tissue-specific activity. To understand how SORT nanoparticles surpass the delivery barrier of liver hepatocyte accumulation, we studied the mechanistic factors which define their organ-targeting properties. We discovered that the chemical nature of the added SORT molecule controlled biodistribution, global/apparent pKa, and serum protein interactions of SORT nanoparticles. Additionally, we provide evidence for an endogenous targeting mechanism whereby organ targeting occurs via 1) desorption of poly(ethylene glycol) lipids from the LNP surface, 2) binding of distinct proteins to the nanoparticle surface because of recognition of exposed SORT molecules, and 3) subsequent interactions between surface-bound proteins and cognate receptors highly expressed in specific tissues. These findings establish a crucial link between the molecular composition of SORT nanoparticles and their unique and precise organ-targeting properties and suggest that the recruitment of specific proteins to a nanoparticle's surface can enable drug delivery beyond the liver.


Subject(s)
Gene Editing/methods , Liposomes , Nanoparticle Drug Delivery System , Nanoparticles , RNA, Messenger , Animals , Humans , Liposomes/metabolism , Liposomes/pharmacokinetics , Liver/metabolism , Mice , Mice, Inbred C57BL , Nanoparticles/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Messenger/pharmacokinetics , Tissue Distribution
5.
J Am Chem Soc ; 145(44): 24302-24314, 2023 11 08.
Article in English | MEDLINE | ID: mdl-37853662

ABSTRACT

Lipid nanoparticles (LNPs) represent the most clinically advanced nonviral mRNA delivery vehicles; however, the full potential of the LNP platform is greatly hampered by inadequate endosomal escape capability. Herein, we rationally introduce a disulfide bond-bridged ester linker to modularly synthesize a library of 96 linker-degradable ionizable lipids (LDILs) for improved mRNA delivery in vivo. The top-performing LDILs are composed of one 4A3 amino headgroup, four disulfide bond-bridged linkers, and four 10-carbon tail chains, whose unique GSH-responsive cone-shaped architectures endow optimized 4A3-SCC-10 and 4A3-SCC-PH lipids with superior endosomal escape and rapid mRNA release abilities, outperforming their parent lipids 4A3-SC-10/PH without a disulfide bond and control lipids 4A3-SSC-10/PH with a disulfide bond in the tail. Notably, compared to DLin-MC3-DMA via systematic administration, 4A3-SCC-10- and 4A3-SCC-PH-formulated LNPs significantly improved mRNA delivery in livers by 87-fold and 176-fold, respectively. Moreover, 4A3-SCC-PH LNPs enabled the highly efficient gene editing of 99% hepatocytes at a low Cre mRNA dose in tdTomato mice following intravenous administration. Meanwhile, 4A3-SCC-PH LNPs were able to selectively deliver firefly luciferase mRNA and facilitate luciferase expression in tumor cells after intraperitoneal injection, further improving cancer metastasis delineation and surgery via bioluminescence imaging. We envision that the chemistry adopted here can be further extended to develop new biodegradable ionizable lipids for broad applications such as gene editing and cancer immunotherapy.


Subject(s)
Nanoparticles , Neoplasms , Mice , Animals , RNA, Messenger/metabolism , Lipids/chemistry , Drug Delivery Systems , Liver/metabolism , Nanoparticles/chemistry , Disulfides/metabolism , RNA, Small Interfering/genetics , Neoplasms/metabolism
6.
Angew Chem Int Ed Engl ; 62(44): e202310395, 2023 10 26.
Article in English | MEDLINE | ID: mdl-37651468

ABSTRACT

Chimeric Antigen Receptor (CAR) T cell immunotherapy is revolutionizing treatment for patients suffering from B-cell lymphoma (BL). However, the current method of CAR T cell production is complicated and expensive, requiring collection of patient blood to enrich the T cell population, ex vivo engineering/activation, and quality assessment before the patient can receive the treatment. Herein we leverage Spleen Selective ORgan Targeted (SORT) Lipid Nanoparticles (LNPs) to produce CAR T cells in situ and bypass the extensive and laborious process currently used. Optimized Spleen SORT LNPs containing 10 % 18 : 1 PA transfected CD3+, CD8+, and CD4+ T cells in wild-type mice. Spleen SORT LNPs delivered Cre recombinase mRNA and CAR encoding mRNA to T cells in reporter mice and in a lymphoreplete B cell lymphoma model (respectively) after intravenous injection without the need for active targeting ligands. Moreover, in situ CAR T cells increased the overall survival of mice with a less aggressive form of B cell lymphoma. In addition, in situ transfected CAR T cells reduced tumor metastasis to the liver by increasing tumor infiltrating lymphocytes. Overall, these results offer a promising alternative method for CAR T cell production with pre-clinical potential to treat hematological malignancies.


Subject(s)
Lymphoma, B-Cell , Receptors, Chimeric Antigen , Humans , Animals , Mice , Spleen , Cell Line, Tumor , Lymphoma, B-Cell/drug therapy , RNA, Messenger
7.
Nat Mater ; 20(5): 701-710, 2021 05.
Article in English | MEDLINE | ID: mdl-33542471

ABSTRACT

Endosomal escape remains a fundamental barrier hindering the advancement of nucleic acid therapeutics. Taking inspiration from natural phospholipids that comprise biological membranes, we report the combinatorial synthesis of multi-tailed ionizable phospholipids (iPhos) capable of delivering messenger RNA or mRNA/single-guide RNA for gene editing in vivo. Optimized iPhos lipids are composed of one pH-switchable zwitterion and three hydrophobic tails, which adopt a cone shape in endosomal acidic environments to facilitate membrane hexagonal transformation and subsequent cargo release from endosomes. Structure-activity relationships reveal that iPhos chemical structure can control in vivo efficacy and organ selectivity. iPhos lipids synergistically function with various helper lipids to formulate multi-component lipid nanoparticles (called iPLNPs) for selective organ targeting. Zwitterionic, ionizable cationic and permanently cationic helper lipids enable tissue-selective mRNA delivery and CRISPR-Cas9 gene editing in spleen, liver and lungs (respectively) following intravenous administration. This rational design of functional phospholipids demonstrates substantial value for gene editing research and therapeutic applications.


Subject(s)
CRISPR-Cas Systems , Cell Membrane/metabolism , Drug Delivery Systems , Gene Editing , Phospholipids , RNA, Messenger , Administration, Intravenous , Animals , Cell Line , Female , Mice , Organ Specificity , Phospholipids/chemistry , Phospholipids/pharmacology , RNA, Messenger/chemistry , RNA, Messenger/pharmacology
8.
Mol Pharm ; 19(11): 3973-3986, 2022 11 07.
Article in English | MEDLINE | ID: mdl-36154076

ABSTRACT

Within the field of lipid nanoparticles (LNPs) for RNA delivery, the focus has been mainly placed on organ level delivery, which can mask cellular level effects consequential to therapeutic applications. Here, we studied a pair of LNPs with similar physical properties and discovered how the chemistry of the ionizable amino lipid can control the endogenous LNP identity, affecting cellular uptake in the liver and altering therapeutic outcomes in a model of liver cancer. Although most LNPs accumulate in the liver after intravenous administration (suggesting that liver delivery is straightforward), we observed an unexpected behavior when comparing two similar LNP formulations (5A2-SC8 and 3A5-SC14 LNPs) that resulted in distinct RNA delivery within the organ. Despite both LNPs possessing similar physical properties, ability to silence gene expression in vitro, strong accumulation within the liver, and a shared pKa of 6.5, only 5A2-SC8 LNPs were able to functionally deliver RNA to hepatocytes. Factor VII (FVII) activity was reduced by 87%, with 5A2-SC8 LNPs carrying FVII siRNA (siFVII), while 3A5-SC14 LNPs carrying siFVII produced baseline FVII activity levels comparable to the nontreatment control at a dosage of 0.5 mg/kg. Protein corona analysis indicated that 5A2-SC8 LNPs bind apolipoprotein E (ApoE), which can drive LDL-R receptor-mediated endocytosis in hepatocytes. In contrast, the surface of 3A5-SC14 LNPs was enriched in albumin but depleted in ApoE, which likely led to Kupffer cell delivery and detargeting of hepatocytes. In an aggressive MYC-driven liver cancer model relevant to hepatocytes, 5A2-SC8 LNPs carrying let-7g miRNA were able to significantly extend survival up to 121 days. Since disease targets exist in an organ- and cell-specific manner, the clinical development of RNA LNP therapeutics will require an improved understanding of LNP cellular tropism within organs. The results from our work illustrate the importance of understanding the cellular localization of RNA delivery and incorporating further checkpoints when choosing nanoparticles beyond biochemical and physical characterization, as small changes in the chemical composition of LNPs can have an impact on both the biofate of LNPs and therapeutic outcomes.


Subject(s)
Liver Neoplasms , Nanoparticles , Humans , Lipids/chemistry , Nanoparticles/chemistry , RNA, Small Interfering , Apolipoproteins E , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Treatment Outcome
9.
J Am Chem Soc ; 143(50): 21321-21330, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34878786

ABSTRACT

Polymers represent a promising therapeutic platform for extrahepatic messenger RNA (mRNA) delivery but are hampered by low in vivo efficacy due to polyplex serum instability and inadequate endosomal escape following systemic administration. Here, we report the rational design and combinatorial synthesis of zwitterionic phospholipidated polymers (ZPPs) via cationic polymer postmodification by alkylated dioxaphospholane oxides to deliver mRNA to spleen and lymph nodes in vivo. This modular postmodification approach readily produces tunable zwitterionic species for serum resistance and introduces alkyl chains simultaneously to enhance endosomal escape, thereby transforming deficient cationic polymers to efficacious zwitterionic mRNA carriers without the need to elaborately synthesize functional monomers. ZPPs mediated up to 39 500-fold higher protein expression than their parent cationic counterparts in vitro and enabled efficacious mRNA delivery selectively in spleen and lymph nodes following intravenous administration in vivo. This zwitterionic phospholipidation methodology provides a versatile and generalizable postmodification strategy to introduce zwitterions into the side chains of cationic polymers, extending the utility of cationic polymer families for precise mRNA delivery and demonstrating substantial potential for immunotherapeutic applications.


Subject(s)
Lymph Nodes/metabolism , Phospholipids/chemistry , Polymers/chemistry , RNA, Messenger/metabolism , Spleen/metabolism , Animals , Cations/chemistry , Endosomes/metabolism , Gene Transfer Techniques , Mice , Mice, Inbred C57BL , RNA, Messenger/chemistry
10.
Proc Natl Acad Sci U S A ; 115(37): E8698-E8706, 2018 09 11.
Article in English | MEDLINE | ID: mdl-30150374

ABSTRACT

Successful cancer immunotherapy entails activation of innate immune receptors to promote dendritic cell (DC) maturation, antigen presentation, up-regulation of costimulatory molecules, and cytokine secretion, leading to activation of tumor antigen-specific cytotoxic T lymphocytes (CTLs). Here we screened a synthetic library of 100,000 compounds for innate immune activators using TNF production by THP-1 cells as a readout. We identified and optimized a potent human and mouse Toll-like receptor (TLR)1/TLR2 agonist, Diprovocim, which exhibited an EC50 of 110 pM in human THP-1 cells and 1.3 nM in primary mouse peritoneal macrophages. In mice, Diprovocim-adjuvanted ovalbumin immunization promoted antigen-specific humoral and CTL responses and synergized with anti-PD-L1 treatment to inhibit tumor growth, generating long-term antitumor memory, curing or prolonging survival of mice engrafted with the murine melanoma B16-OVA. Diprovocim induced greater frequencies of tumor-infiltrating leukocytes than alum, of which CD8 T cells were necessary for the antitumor effect of immunization plus anti-PD-L1 treatment.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antibodies, Monoclonal/pharmacology , B7-H1 Antigen/antagonists & inhibitors , Melanoma, Experimental/therapy , Toll-Like Receptor 1/agonists , Toll-Like Receptor 2/agonists , Animals , Antibodies, Monoclonal/immunology , B7-H1 Antigen/immunology , Cell Line, Tumor , Cells, Cultured , Drug Synergism , Humans , Immunotherapy/methods , Kaplan-Meier Estimate , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology , THP-1 Cells , Toll-Like Receptor 1/genetics , Toll-Like Receptor 1/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism
11.
Angew Chem Int Ed Engl ; 60(11): 5848-5853, 2021 03 08.
Article in English | MEDLINE | ID: mdl-33305471

ABSTRACT

Lipid nanoparticles (LNPs) represent the leading concept for mRNA delivery. Unsaturated lipids play important roles in nature with potential for mRNA therapeutics, but are difficult to access through chemical synthesis. To systematically study the role of unsaturation, modular reactions were utilized to access a library of 91 amino lipids, enabled by the synthesis of unsaturated thiols. An ionizable lipid series (4A3) emerged from in vitro and in vivo screening, where the 4A3 core with a citronellol-based (Cit) periphery emerged as best. We studied the interaction between LNPs and a model endosomal membrane where 4A3-Cit demonstrated superior lipid fusion over saturated lipids, suggesting its unsaturated tail promotes endosomal escape. Furthermore, 4A3-Cit significantly improved mRNA delivery efficacy in vivo through Selective ORgan Targeting (SORT), resulting in 18-fold increased protein expression over parent LNPs. These findings provide insight into how lipid unsaturation promotes mRNA delivery and demonstrate how lipid mixing can enhance efficacy.


Subject(s)
Lipids/chemistry , Nanoparticles/chemistry , RNA, Messenger/genetics , Animals , Endosomes/chemistry , Endosomes/metabolism , Gene Transfer Techniques , Lipids/administration & dosage , Mice , Mice, Inbred C57BL , Molecular Structure , Nanoparticles/administration & dosage , Nanoparticles/metabolism , RNA, Messenger/administration & dosage , RNA, Messenger/chemistry
12.
Mol Pharm ; 17(5): 1575-1585, 2020 05 04.
Article in English | MEDLINE | ID: mdl-32267707

ABSTRACT

In this work, a series of linear-dendritic poly(ethylene glycol) (PEG) lipids (PEG-GnCm) were synthesized through a strategy using sequential aza- and sulfa-Michael addition reactions. The effect of modulating the hydrophobic domain of linear-dendritic PEG lipids was systematically investigated for in vitro and in vivo small RNA delivery as the surface-stabilizing component of 5A2-SC8 dendrimer lipid-based nanoparticles (DLNPs). The lipid alkyl lengths (C8, C12, and C16) and dendrimer generations (G1, G2, and G3) were altered to create PEG-GnCm with different physical properties and anchoring potential. The tail chemical structure of PEG-GnCm did not affect the formulation of 5A2-SC8 DLNPs, including the nanoparticle size, RNA encapsulation, and stability. However, the tail chemical structure did dramatically affect the RNA delivery efficacy of the formed 5A2-SC8 DLNPs with different PEG-GnCm. First-generation PEG lipids (PEG-G1C8, PEG-G1C12, and PEG-G1C16) and a second-generation PEG lipid (PEG-G2C8) formed 5A2-SC8 DLNPs that could deliver siRNAs effectively in vitro and in vivo. 5A2-SC8 DLNPs formulated with second-generation PEG lipids (PEG-G2C12 and PEG-G2C16) and all three third-generation PEG lipids (PEG-G3C8, PEG-G3C12, and PEG-G3C16) lost the ability to deliver siRNA effectively in vitro and in vivo. Overall, we determined that the hydrophobic domain chemical structure of linear-dendritic poly(ethylene glycol) lipids affected the RNA delivery of DLNPs by impacting the escape of 5A2-SC8 DLNPs from endosomes at early cell incubation times, thereby indicating how PEG lipid anchoring and chemical structure can modulate in vitro and in vivo siRNA delivery efficacies.


Subject(s)
Dendrimers/chemistry , Drug Delivery Systems , Lipids/chemistry , Nanoparticles/chemistry , Polyethylene Glycols/chemistry , RNA, Small Interfering/administration & dosage , Animals , HeLa Cells , Humans , Hydrophobic and Hydrophilic Interactions , Mice , Mice, Inbred C57BL , RNA, Small Interfering/chemistry
13.
Proc Natl Acad Sci U S A ; 114(3): 580-585, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28049847

ABSTRACT

Macrophages are prominent immune cells in the tumor microenvironment that exert potent effects on cancer metastasis. However, the signals and receivers for the tumor-macrophage communication remain enigmatic. Here, we show that G protein-coupled receptor 132 (Gpr132) functions as a key macrophage sensor of the rising lactate in the acidic tumor milieu to mediate the reciprocal interaction between cancer cells and macrophages during breast cancer metastasis. Lactate activates macrophage Gpr132 to promote the alternatively activated macrophage (M2)-like phenotype, which, in turn, facilitates cancer cell adhesion, migration, and invasion. Consequently, Gpr132 deletion reduces M2 macrophages and impedes breast cancer lung metastasis in mice. Clinically, Gpr132 expression positively correlates with M2 macrophages, metastasis, and poor prognosis in patients with breast cancer. These findings uncover the lactate-Gpr132 axis as a driver of breast cancer metastasis by stimulating tumor-macrophage interplay, and reveal potential new therapeutic targets for breast cancer treatment.


Subject(s)
Breast Neoplasms/metabolism , Cell Cycle Proteins/metabolism , Lactic Acid/metabolism , Macrophages/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Adhesion , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Movement , Female , Humans , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Macrophage Activation , Macrophages/immunology , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Invasiveness , Prognosis , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Tumor Microenvironment
14.
Gastroenterology ; 154(5): 1421-1434, 2018 04.
Article in English | MEDLINE | ID: mdl-29274368

ABSTRACT

BACKGROUND & AIMS: Cytokinesis can fail during normal postnatal liver development, leading to polyploid hepatocytes. We investigated whether inhibiting cytokinesis in the liver slows tumor growth without compromising the health of normal hepatocytes. We inhibited cytokinesis in cancer cells by knocking down ANLN, a cytoskeletal scaffolding protein that regulates cytokinesis and might promote tumorigenesis, in mice with liver disease. METHODS: We analyzed clinical and gene expression data from The Cancer Genome Atlas, Oncomine, PrognoScan, and a hepatocellular carcinoma (HCC) tissue microarray. We knocked down ANLN with small interfering RNAs (siRNAs) in H2.35 liver cells and performed image analyses of cells undergoing cytokinesis. siRNAs were delivered to LAP-MYC mice, which develop hepatoblastoma, using lipid nanoparticles. H2.35 cells with knockdown of ANLN or control cells were injected into FRG mice, which develop chronic liver damage, and tumor growth was monitored. We also developed mice with inducible expression of transgenes encoding small hairpin RNAs (shRNAs) against Anln messenger RNA and studied liver tumorigenesis after administration of diethylnitrosamine and carbon tetrachloride. siRNAs against Anln messenger RNA were conjugated to N-acetylgalactosamine to reduce toxicity and increase hepatocyte tropism; their effects were studied in mouse models of liver cancer and regeneration. RESULTS: Levels of ANLN messenger RNA were increased in human HCC tissues compared to non-tumor liver tissues. siRNA knockdown of ANLN blocked cytokinesis in H2.35 liver cells. Administration of siRNA against ANLN increased survival times of LAP-MYC mice, compared to mice given a control siRNA. H2.35 liver cells with shRNA knockdown of ANLN formed tumors more slowly in FRG mice than control H2.35 cells. Mice with inducible expression of shRNAs against Anln mRNA developed fewer liver tumors after administration of diethylnitrosamine and carbon tetrachloride than control mice. Knockdown of ANLN did not affect liver regeneration after acute and chronic liver injuries. CONCLUSIONS: Knockdown of ANLN in liver cells blocks cytokinesis and inhibits development of liver tumors in mice. Agents that inhibit ANLN in the liver might be effective for prevention or treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carrier Proteins/genetics , Cell Transformation, Neoplastic/metabolism , Cytokinesis , Hepatocytes/metabolism , Liver Neoplasms/metabolism , Liver Regeneration , Microfilament Proteins/deficiency , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/prevention & control , Cell Line , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Genetic Predisposition to Disease , Hepatectomy , Hepatocytes/pathology , Hepatocytes/transplantation , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/prevention & control , Mice, Knockout , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Phenotype , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , Time Factors , Transfection
15.
Proc Natl Acad Sci U S A ; 113(3): 520-5, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26729861

ABSTRACT

RNA-based cancer therapies are hindered by the lack of delivery vehicles that avoid cancer-induced organ dysfunction, which exacerbates carrier toxicity. We address this issue by reporting modular degradable dendrimers that achieve the required combination of high potency to tumors and low hepatotoxicity to provide a pronounced survival benefit in an aggressive genetic cancer model. More than 1,500 dendrimers were synthesized using sequential, orthogonal reactions where ester degradability was systematically integrated with chemically diversified cores, peripheries, and generations. A lead dendrimer, 5A2-SC8, provided a broad therapeutic window: identified as potent [EC50 < 0.02 mg/kg siRNA against FVII (siFVII)] in dose-response experiments, and well tolerated in separate toxicity studies in chronically ill mice bearing MYC-driven tumors (>75 mg/kg dendrimer repeated dosing). Delivery of let-7 g microRNA (miRNA) mimic inhibited tumor growth and dramatically extended survival. Efficacy stemmed from a combination of a small RNA with the dendrimer's own negligible toxicity, therefore illuminating an underappreciated complication in treating cancer with RNA-based drugs.


Subject(s)
Dendrimers/chemistry , Liver Neoplasms/pathology , Models, Biological , RNA, Small Interfering/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Dendrimers/toxicity , Drug Carriers/chemistry , Drug Delivery Systems , Esters/chemistry , Extracellular Space/chemistry , Fluorescence , HeLa Cells , Humans , Intracellular Space/chemistry , Mice , MicroRNAs/metabolism , Molecular Weight , Nanoparticles/chemistry , Proto-Oncogene Proteins c-myc/metabolism
16.
Proc Natl Acad Sci U S A ; 113(39): E5702-10, 2016 09 27.
Article in English | MEDLINE | ID: mdl-27621434

ABSTRACT

Conventional chemotherapeutics nonselectively kill all rapidly dividing cells, which produces numerous side effects. To address this challenge, we report the discovery of functional polyesters that are capable of delivering siRNA drugs selectively to lung cancer cells and not to normal lung cells. Selective polyplex nanoparticles (NPs) were identified by high-throughput library screening on a unique pair of matched cancer/normal cell lines obtained from a single patient. Selective NPs promoted rapid endocytosis into HCC4017 cancer cells, but were arrested at the membrane of HBEC30-KT normal cells during the initial transfection period. When injected into tumor xenografts in mice, cancer-selective NPs were retained in tumors for over 1 wk, whereas nonselective NPs were cleared within hours. This translated to improved siRNA-mediated cancer cell apoptosis and significant suppression of tumor growth. Selective NPs were also able to mediate gene silencing in xenograft and orthotopic tumors via i.v. injection or aerosol inhalation, respectively. Importantly, this work highlights that different cells respond differentially to the same drug carrier, an important factor that should be considered in the design and evaluation of all NP carriers. Because no targeting ligands are required, these functional polyester NPs provide an exciting alternative approach for selective drug delivery to tumor cells that may improve efficacy and reduce adverse side effects of cancer therapies.


Subject(s)
Gene Transfer Techniques , Lung Neoplasms/therapy , Polyesters/chemistry , RNA, Small Interfering/metabolism , Animals , Apoptosis , Carbocyanines , Cell Line, Tumor , Cell Proliferation , Combinatorial Chemistry Techniques , Endocytosis , Gene Silencing , Humans , Mice , Nanoparticles/chemistry , Ubiquitin/metabolism , Xenograft Model Antitumor Assays
17.
Biomacromolecules ; 18(12): 4307-4315, 2017 Dec 11.
Article in English | MEDLINE | ID: mdl-29141136

ABSTRACT

Messenger RNA (mRNA) has recently come into focus as an emerging therapeutic class with great potential for protein replacement therapy, cancer immunotherapy, regenerative medicine, vaccines, and gene editing. However, the lack of effective and safe delivery methods impedes the broad application of mRNA-based therapeutics. We report a robust approach to develop efficient polymeric delivery carriers for mRNA. Lead polyesters were identified by in vitro screening of a 480-member combinatorially modified poly(trimethylolpropane allyl ether-co-suberoyl chloride) library for the delivery of luciferase encoding mRNA (Luc mRNA) to IGROV1 cells. The formulation of mRNA polyplex nanoparticles (NPs) with Pluronic F127 decreased the surface charge. Although this improved the stability of mRNA nanoparticles, the delivery potency decreased with increased F127 content. Thus, we determined that NP stabilization with 5% F127 could balance the protective effects and delivery potency. 5% F127 formulated PE4K-A17-0.33C12 mRNA NPs enabled luciferase expression predominantly in the lungs after intravenous injection into mice. The efficient mRNA delivery specifically to lungs by degradable carriers suggests the potential for the treatment of pulmonary diseases.


Subject(s)
Drug Carriers/chemistry , Lung/drug effects , Polyesters/chemistry , RNA, Messenger/administration & dosage , RNA, Messenger/chemistry , Animals , Cell Line, Tumor , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Nanoparticles/chemistry , Poloxamer/chemistry , Polymers/chemistry
18.
Proc Natl Acad Sci U S A ; 111(11): 3955-60, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24516150

ABSTRACT

siRNA therapeutics have promise for the treatment of a wide range of genetic disorders. Motivated by lipoproteins, we report lipopeptide nanoparticles as potent and selective siRNA carriers with a wide therapeutic index. Lead material cKK-E12 showed potent silencing effects in mice (ED50 ∼ 0.002 mg/kg), rats (ED50 < 0.01 mg/kg), and nonhuman primates (over 95% silencing at 0.3 mg/kg). Apolipoprotein E plays a significant role in the potency of cKK-E12 both in vitro and in vivo. cKK-E12 was highly selective toward liver parenchymal cell in vivo, with orders of magnitude lower doses needed to silence in hepatocytes compared with endothelial cells and immune cells in different organs. Toxicity studies showed that cKK-E12 was well tolerated in rats at a dose of 1 mg/kg (over 100-fold higher than the ED50). To our knowledge, this is the most efficacious and selective nonviral siRNA delivery system for gene silencing in hepatocytes reported to date.


Subject(s)
Drug Delivery Systems/methods , Lipopeptides/chemistry , Nanoparticles/chemistry , RNA, Small Interfering/administration & dosage , Animals , Apolipoproteins E/metabolism , Cryoelectron Microscopy , Gene Silencing , Hepatocytes/metabolism , Macaca fascicularis , Mice , RNA, Small Interfering/therapeutic use , Rats
19.
Angew Chem Int Ed Engl ; 56(4): 1059-1063, 2017 01 19.
Article in English | MEDLINE | ID: mdl-27981708

ABSTRACT

CRISPR/Cas is a revolutionary gene editing technology with wide-ranging utility. The safe, non-viral delivery of CRISPR/Cas components would greatly improve future therapeutic utility. We report the synthesis and development of zwitterionic amino lipids (ZALs) that are uniquely able to (co)deliver long RNAs including Cas9 mRNA and sgRNAs. ZAL nanoparticle (ZNP) delivery of low sgRNA doses (15 nm) reduces protein expression by >90 % in cells. In contrast to transient therapies (such as RNAi), we show that ZNP delivery of sgRNA enables permanent DNA editing with an indefinitely sustained 95 % decrease in protein expression. ZNP delivery of mRNA results in high protein expression at low doses in vitro (<600 pM) and in vivo (1 mg kg-1 ). Intravenous co-delivery of Cas9 mRNA and sgLoxP induced expression of floxed tdTomato in the liver, kidneys, and lungs of engineered mice. ZNPs provide a chemical guide for rational design of long RNA carriers, and represent a promising step towards improving the safety and utility of gene editing.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Editing , Gene Transfer Techniques , Nanoparticles/chemistry , RNA, Guide, Kinetoplastida/genetics , RNA, Messenger/genetics , Lipids/chemistry , Molecular Structure , RNA, Guide, Kinetoplastida/chemistry , RNA, Messenger/chemistry
20.
Bioconjug Chem ; 27(7): 1737-44, 2016 Jul 20.
Article in English | MEDLINE | ID: mdl-27285307

ABSTRACT

Dysregulated pH has been recognized as a universal tumor microenvironment signature that can delineate tumors from normal tissues. Existing fluorescent probes that activate in response to pH are hindered by either fast clearance (in the case of small molecules) or high liver background emission (in the case of large particles). There remains a need to design water-soluble, long circulating, pH-responsive nanoprobes with high tumor-to-liver contrast. Herein, we report a modular chemical strategy to create acidic pH-sensitive and water-soluble fluorescent probes for high in vivo tumor detection and minimal liver activation. A combination of a modified Knoevenagel reaction and PEGylation yielded a series of NIR BODIPY fluorophores with tunable pKas, high quantum yield, and optimal orbital energies to enable photoinduced electron transfer (PeT) activation in response to pH. After intravenous administration, Probe 5c localized to tumors and provided excellent tumor-to-liver contrast (apparent T/L = 3) because it minimally activates in the liver. This phenomenon was further confirmed by direct ex vivo imaging experiments on harvested organs. Because no targeting ligands were required, we believe that this report introduces a versatile strategy to directly synthesize soluble probes with broad potential utility including fluorescence-based image-guided surgery, cancer diagnosis, and theranostic nanomedicine.


Subject(s)
Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Liver/metabolism , Mammary Neoplasms, Experimental/metabolism , Water/chemistry , Animals , Boron Compounds/chemistry , Cell Line, Tumor , Humans , Hydrogen-Ion Concentration , Mice , Molecular Weight , Solubility , Spectrometry, Fluorescence
SELECTION OF CITATIONS
SEARCH DETAIL