Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
2.
Nat Commun ; 12(1): 4219, 2021 07 09.
Article in English | MEDLINE | ID: mdl-34244505

ABSTRACT

Streptococcus pyogenes (Spy) Cas9 has potential as a component of gene therapeutics for incurable diseases. One of its limitations is its large size, which impedes its formulation and delivery in therapeutic applications. Smaller Cas9s are an alternative, but lack robust activity or specificity and frequently recognize longer PAMs. Here, we investigated four uncharacterized, smaller Cas9s and found three employing a "GG" dinucleotide PAM similar to SpyCas9. Protein engineering generated synthetic RNA-guided nucleases (sRGNs) with editing efficiencies and specificities exceeding even SpyCas9 in vitro and in human cell lines on disease-relevant targets. sRGN mRNA lipid nanoparticles displayed manufacturing advantages and high in vivo editing efficiency in the mouse liver. Finally, sRGNs, but not SpyCas9, could be packaged into all-in-one AAV particles with a gRNA and effected robust in vivo editing of non-human primate (NHP) retina photoreceptors. Human gene therapy efforts are expected to benefit from these improved alternatives to existing CRISPR nucleases.


Subject(s)
CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems/genetics , Gene Editing/methods , Staphylococcus/enzymology , Animals , CRISPR-Associated Protein 9/isolation & purification , Cell Line, Tumor , Dependovirus , Disease Models, Animal , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , HEK293 Cells , Humans , Macaca fascicularis , Male , Mice , Parvovirinae/genetics , Protein Engineering , Ribonucleases , Staphylococcus/genetics , Substrate Specificity , Usher Syndromes/genetics , Usher Syndromes/therapy , RNA, Guide, CRISPR-Cas Systems
3.
Nature ; 593(7859): 429-434, 2021 05.
Article in English | MEDLINE | ID: mdl-34012082

ABSTRACT

Gene-editing technologies, which include the CRISPR-Cas nucleases1-3 and CRISPR base editors4,5, have the potential to permanently modify disease-causing genes in patients6. The demonstration of durable editing in target organs of nonhuman primates is a key step before in vivo administration of gene editors to patients in clinical trials. Here we demonstrate that CRISPR base editors that are delivered in vivo using lipid nanoparticles can efficiently and precisely modify disease-related genes in living cynomolgus monkeys (Macaca fascicularis). We observed a near-complete knockdown of PCSK9 in the liver after a single infusion of lipid nanoparticles, with concomitant reductions in blood levels of PCSK9 and low-density lipoprotein cholesterol of approximately 90% and about 60%, respectively; all of these changes remained stable for at least 8 months after a single-dose treatment. In addition to supporting a 'once-and-done' approach to the reduction of low-density lipoprotein cholesterol and the treatment of atherosclerotic cardiovascular disease (the leading cause of death worldwide7), our results provide a proof-of-concept for how CRISPR base editors can be productively applied to make precise single-nucleotide changes in therapeutic target genes in the liver, and potentially in other organs.


Subject(s)
CRISPR-Cas Systems , Cholesterol, LDL/blood , Gene Editing , Models, Animal , Proprotein Convertase 9/genetics , Adenine/metabolism , Animals , Cells, Cultured , Female , Hepatocytes/metabolism , Humans , Liver/enzymology , Loss of Function Mutation , Macaca fascicularis/blood , Macaca fascicularis/genetics , Male , Mice , Mice, Inbred C57BL , Mutagenesis, Site-Directed , Proprotein Convertase 9/blood , Proprotein Convertase 9/metabolism , Time Factors
4.
Oncogene ; 38(12): 2151-2161, 2019 03.
Article in English | MEDLINE | ID: mdl-30459357

ABSTRACT

MicroRNAs (miRNAs) have emerged as crucial players in the development and maintenance of disease. miR-155 is an inflammation-associated, oncogenic miRNA, frequently overexpressed in hematological malignancies and solid tumors. However, the mechanism of oncogenesis by miR-155 is not well characterized, and research has focused primarily on individual, direct targets, which does not recapitulate the complexities of cancer. Using a powerful, inducible transgenic mouse model that overexpresses miR-155 and develops miR-155-addicted hematological malignancy, we describe here a multi-step process of oncogenesis by miR-155, which involves cooperation between miR-155, its direct targets, and other oncogenes. miR-155 is known to target DNA-repair proteins, leading to a mutator phenotype, and we find that over 93% of tumors in our miR-155 overexpressing mice contain activating mutations in a single oncogene, c-Kit. Treating mice with dasatinib or imatinib, which target c-Kit, resulted in complete tumor regression, indicating that c-Kit activity is crucial in the oncogenic process. Interestingly, c-Kit expression is high when miR-155 is overexpressed, indicating further cooperation between miR-155 and c-Kit. Our findings support a multi-step model of oncogenesis by miR-155 in which miR-155 promotes both a mutator phenotype and a cellular environment particularly susceptible to mutations in a given oncogene.


Subject(s)
Carcinogenesis/genetics , MicroRNAs/genetics , Mutation , Oncogenes/genetics , Proto-Oncogene Proteins c-kit/genetics , Animals , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Mice
5.
RNA Biol ; 15(7): 914-922, 2018.
Article in English | MEDLINE | ID: mdl-29923457

ABSTRACT

Arginase I (ARG1) deficiency is an autosomal recessive urea cycle disorder, caused by deficiency of the enzyme Arginase I, resulting in accumulation of arginine in blood. Current Standard of Care (SOC) for ARG1 deficiency in patients or those having detrimental mutations of ARG1 gene is diet control. Despite diet and drug therapy with nitrogen scavengers, ~25% of patients suffer from severe mental deficits and loss of ambulation. 75% of patients whose symptoms can be managed through diet therapy continue to suffer neuro-cognitive deficits. In our research, we demonstrate in vitro and in vivo that administration of ARG1 mRNA increased ARG1 protein expression and specific activity in relevant cell types, including ARG1-deficient patient cell lines, as well as in wild type mice for up to 4 days. These studies demonstrate that ARG1 mRNA treatment led to increased functional protein expression of ARG1 and subsequently an increase in urea. Hence, ARG1 mRNA therapy could be a potential treatment option to develop for patients.


Subject(s)
Arginase/metabolism , Arginine/metabolism , Biological Therapy/methods , Hyperargininemia/therapy , RNA, Messenger/administration & dosage , Animals , Arginase/genetics , HeLa Cells , Hep G2 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mutation , Urea/metabolism
6.
Biomaterials ; 176: 122-130, 2018 09.
Article in English | MEDLINE | ID: mdl-29879653

ABSTRACT

Gene delivery is known to be a complicated multi-step biological process. It has been observed that subtle differences in the structure and properties of polymeric materials used for gene delivery can lead to dramatic differences in transfection efficiency. Therefore, screening of properties is pivotal to optimizing the polymer. So far, most polymeric materials are built in a "bottom-up" manner, i.e. synthesized from monomers that allow modification of polymer composition or structural factors. With this method, we previously synthesized and screened a library of biodegradable poly(amine-co-ester) (PACE) terpolymers for optimized DNA delivery. However, it can be tedious and time consuming to synthesize a polymer library for screening, particularly when small changes of a factor need to be tested, when multiple factors are involved, and when the effects of different factors are synergistic. In the present work, we evaluate the potential of PACE to deliver mRNA. After observing that mRNA transfection efficiency was highly dependent on both end group composition and molecular weight (MW) of PACE in a synergistic manner, we developed a "top-down" process we called actuation, to simultaneously vary these two factors. Some of the actuated PACE (aPACE) materials presented superior mRNA delivery properties compared to regular PACE, with up to a 106-fold-increase in mRNA transfection efficiency in vitro. Moreover, when aPACE was used to deliver mRNA coding for erythropoietin (EPO) in vivo, it produced high levels of EPO in the blood for up to 48 h without inducing systemic toxicity. This polymer constitutes a new delivery vehicle for mRNA-based treatments that provides safe yet potent protein production.


Subject(s)
Polyamines/chemistry , Polyesters/chemistry , RNA, Messenger/administration & dosage , Animals , Cell Line , Cell Survival , Erythropoietin/genetics , Female , Gene Transfer Techniques , Humans , Hydrolysis , Hydrophobic and Hydrophilic Interactions , Mice, Inbred BALB C , Molecular Weight , Nanoparticles/chemistry , Polymerization , Small Molecule Libraries , Transfection
7.
Breast Cancer Res Treat ; 170(1): 55-67, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29511965

ABSTRACT

PURPOSE: Obesity and weight gain are associated with comorbidities including a higher risk of tumor recurrence and cancer-related deaths among breast cancer (BC) survivors; however, the underlying mechanisms linking obesity and cancer are poorly understood. Given the lack of clinically validated BC biomarkers, obesity and weight-loss studies utilize serum biomarkers as the intermediary outcomes of tumor recurrence. Studies have indicated microRNAs (miRNA)s are reliable biomarkers for cancer. We hypothesized that miRNA expression correlates with obesity and weight loss amongst BC survivors. This would yield insight into the biological pathways by which this association occurs, enabling more precise development of therapeutics. PATIENTS AND METHODS: We correlated baseline body mass index (BMI) with serum miRNA expression in 121 BC survivors enrolled in the Hormones and Physical Exercise (HOPE) trial. We then analyzed expression of the 35 most abundant miRNAs from HOPE in a six-month randomized controlled weight-loss trial (Lifestyle, Exercise, and Nutrition; LEAN) in 100 BC survivors. Ingenuity pathway analysis (IPA) software was used to identify biological pathway targets of the BMI-associated and intervention-responsive miRNAs using predictive biomarkers. RESULTS: Pearson correlations in HOPE identified eight miRNAs associated with BMI, including miR-191-5p (r = - 0.22, p = 0.016) and miR-122-5p (r = 0.25, p = 0.0048). In the LEAN validation study, levels of miR-191-5p significantly increased during the six-month intervention (p = 0.082). Ingenuity Pathway Analysis identified "Estrogen-mediated S-phase entry" (HOPE p = 0.003; LEAN p < 0.001) and "Molecular mechanisms of cancer" (HOPE p = 0.02; LEAN p < 0.001) as the top canonical pathways that significantly correlated with BMI-associated and intervention-responsive miRNAs and contain obesity and cancer-relevant genes including the E2F family of transcription factors and CCND1, which have been implicated in sporadic BC. CONCLUSION: While the association between obesity and BC recurrence and mortality has been demonstrated in the literature, mechanisms underlying the link between weight gain and cancer are unclear. Using two independent clinical trials, we identified novel miRNAs associative to BMI and weight loss that contribute to the development of cancer. Predictive modeling of miRNA targets identified multiple canonical pathways associated with cancer, highlighting potential mechanisms explaining the link between BMI and increased cancer risk.


Subject(s)
Breast Neoplasms/therapy , Exercise/physiology , Neoplasm Recurrence, Local/prevention & control , Adult , Aged , Biomarkers, Tumor/genetics , Body Mass Index , Breast Neoplasms/complications , Breast Neoplasms/genetics , Breast Neoplasms/physiopathology , Female , Humans , MicroRNAs/genetics , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/physiopathology , Obesity/complications , Obesity/physiopathology , Obesity/therapy , Weight Loss
8.
RNA Biol ; 15(6): 756-762, 2018.
Article in English | MEDLINE | ID: mdl-29578372

ABSTRACT

mRNA based therapies hold great promise for the treatment of genetic diseases. However, this therapeutic approach suffers from multiple challenges including the short half-life of exogenously administered mRNA and subsequent protein production. Modulation of untranslated regions (UTR) represents one approach to enhance both mRNA stability and translation efficiency. The current studies describe and validate screening methods using a diverse set of 5'UTR and 3'UTR combinations for improved expression of the Arginase 1 (ARG1) protein, a potential therapeutic mRNA target. Data revealed a number of critical aspects which need to be considered when developing a screening approach for engineering mRNA improvements. First, plasmid-based screening methods do not correlate with protein expression driven by exogenously expressed mRNA. Second, improved ARG1 protein production was driven by increased translation and not improved mRNA stability. Finally, the 5' UTR appears to be the key driver in protein expression for exogenously delivered mRNA. From the testing of the combinatorial library, the 5'UTR for complement factor 3 (C3) and cytochrome p4502E1 (CYP2E1) showed the largest and most consistent increase in protein expression relative to a reference UTR. Collectively, these data provide important information for the development and optimization of therapeutic mRNAs.


Subject(s)
3' Untranslated Regions , 5' Untranslated Regions , Arginase , Complement C3/genetics , Cytochrome P-450 CYP2E1/genetics , Protein Biosynthesis/genetics , Arginase/biosynthesis , Arginase/genetics
9.
Nucleic Acid Ther ; 28(2): 74-85, 2018 04.
Article in English | MEDLINE | ID: mdl-29437538

ABSTRACT

Messenger RNA (mRNA) is a promising new class of therapeutics that has potential for treatment of diseases in fields such as immunology, oncology, vaccines, and inborn errors of metabolism. mRNA therapy has several advantages over DNA-based gene therapy, including the lack of the need for nuclear import and transcription, as well as limited possibility of genomic integration. One drawback of mRNA therapy, especially in cases such as metabolic disorders where repeated dosing will be necessary, is the relatively short in vivo half-life of mRNA (∼6-12 h). We hypothesize that protein engineering designed to improve translation, yielding longer-lasting protein, or modifications that would increase enzymatic activity would be helpful in alleviating this issue. In this study, we present two examples where sequence engineering improved the expression and duration, as well as enzymatic activity of target proteins in vitro. We then confirmed these findings in wild-type mice. This work shows that rational engineering of proteins can lead to improved therapies in vivo.


Subject(s)
Arginase/genetics , Hyperargininemia/therapy , Hypoxanthine Phosphoribosyltransferase/genetics , Lesch-Nyhan Syndrome/therapy , RNA, Messenger/therapeutic use , Amino Acid Sequence , Animals , Arginase/isolation & purification , Arginase/metabolism , HeLa Cells , Humans , Hyperargininemia/blood , Hypoxanthine Phosphoribosyltransferase/isolation & purification , Hypoxanthine Phosphoribosyltransferase/metabolism , Lesch-Nyhan Syndrome/blood , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Nanoparticles/therapeutic use , Protein Engineering , RNA, Messenger/genetics , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Alignment
10.
Nat Commun ; 7: 13304, 2016 10 26.
Article in English | MEDLINE | ID: mdl-27782131

ABSTRACT

The blood disorder, ß-thalassaemia, is considered an attractive target for gene correction. Site-specific triplex formation has been shown to induce DNA repair and thereby catalyse genome editing. Here we report that triplex-forming peptide nucleic acids (PNAs) substituted at the γ position plus stimulation of the stem cell factor (SCF)/c-Kit pathway yielded high levels of gene editing in haematopoietic stem cells (HSCs) in a mouse model of human ß-thalassaemia. Injection of thalassemic mice with SCF plus nanoparticles containing γPNAs and donor DNAs ameliorated the disease phenotype, with sustained elevation of blood haemoglobin levels into the normal range, reduced reticulocytosis, reversal of splenomegaly and up to 7% ß-globin gene correction in HSCs, with extremely low off-target effects. The combination of nanoparticle delivery, next generation γPNAs and SCF treatment may offer a minimally invasive treatment for genetic disorders of the blood that can be achieved safely and simply by intravenous administration.


Subject(s)
Gene Editing/methods , Genetic Therapy/methods , Hematopoietic Stem Cells/metabolism , Peptide Nucleic Acids/genetics , beta-Thalassemia/therapy , Animals , Cell Line , DNA/administration & dosage , DNA/genetics , Disease Models, Animal , Hemoglobins/analysis , Humans , Injections, Intravenous , Mice , Mice, Transgenic , Nanoparticles/administration & dosage , Peptide Nucleic Acids/administration & dosage , Proto-Oncogene Proteins c-kit/metabolism , Stem Cell Factor/administration & dosage , Stem Cell Factor/metabolism , beta-Globins/genetics , beta-Thalassemia/blood , beta-Thalassemia/genetics
11.
Cancer Res ; 76(4): 927-39, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26676753

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive subtype with no clinically proven biologically targeted treatment options. The molecular heterogeneity of TNBC and lack of high frequency driver mutations other than TP53 have hindered the development of new and effective therapies that significantly improve patient outcomes. miRNAs, global regulators of survival and proliferation pathways important in tumor development and maintenance, are becoming promising therapeutic agents. We performed miRNA-profiling studies in different TNBC subtypes to identify miRNAs that significantly contribute to disease progression. We found that miR-34a was lost in TNBC, specifically within mesenchymal and mesenchymal stem cell-like subtypes, whereas expression of miR-34a targets was significantly enriched. Furthermore, restoration of miR-34a in cell lines representing these subtypes inhibited proliferation and invasion, activated senescence, and promoted sensitivity to dasatinib by targeting the proto-oncogene c-SRC. Notably, SRC depletion in TNBC cell lines phenocopied the effects of miR-34a reintroduction, whereas SRC overexpression rescued the antitumorigenic properties mediated by miR-34a. miR-34a levels also increased when cells were treated with c-SRC inhibitors, suggesting a negative feedback exists between miR-34a and c-SRC. Moreover, miR-34a administration significantly delayed tumor growth of subcutaneously and orthotopically implanted tumors in nude mice, and was accompanied by c-SRC downregulation. Finally, we found that miR-34a and SRC levels were inversely correlated in human tumor specimens. Together, our results demonstrate that miR-34a exerts potent antitumorigenic effects in vitro and in vivo and suggests that miR-34a replacement therapy, which is currently being tested in human clinical trials, represents a promising therapeutic strategy for TNBC.


Subject(s)
MicroRNAs/metabolism , src-Family Kinases/metabolism , Animals , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Proto-Oncogene Mas , Triple Negative Breast Neoplasms/genetics
12.
Proc Natl Acad Sci U S A ; 112(48): E6597-605, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26627251

ABSTRACT

Canonical siRNA design algorithms have become remarkably effective at predicting favorable binding regions within a target mRNA, but in some cases (e.g., a fusion junction site) region choice is restricted. In these instances, alternative approaches are necessary to obtain a highly potent silencing molecule. Here we focus on strategies for rational optimization of two siRNAs that target the junction sites of fusion oncogenes BCR-ABL and TMPRSS2-ERG. We demonstrate that modifying the termini of these siRNAs with a terminal G-U wobble pair or a carefully selected pair of terminal asymmetry-enhancing mismatches can result in an increase in potency at low doses. Importantly, we observed that improvements in silencing at the mRNA level do not necessarily translate to reductions in protein level and/or cell death. Decline in protein level is also heavily influenced by targeted protein half-life, and delivery vehicle toxicity can confound measures of cell death due to silencing. Therefore, for BCR-ABL, which has a long protein half-life that is difficult to overcome using siRNA, we also developed a nontoxic transfection vector: poly(lactic-coglycolic acid) nanoparticles that release siRNA over many days. We show that this system can achieve effective killing of leukemic cells. These findings provide insights into the implications of siRNA sequence for potency and suggest strategies for the design of more effective therapeutic siRNA molecules. Furthermore, this work points to the importance of integrating studies of siRNA design and delivery, while heeding and addressing potential limitations such as restricted targetable mRNA regions, long protein half-lives, and nonspecific toxicities.


Subject(s)
Fusion Proteins, bcr-abl/genetics , Gene Expression Regulation, Leukemic , Gene Targeting/methods , Oncogene Proteins, Fusion/genetics , RNA, Small Interfering/metabolism , Apoptosis , Base Sequence , Binding Sites , Cell Line, Tumor , Cell Survival , Drug Delivery Systems , HEK293 Cells , Humans , K562 Cells , Lactic Acid/chemistry , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Nanoparticles , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , RNA Interference , RNA, Messenger/metabolism , Transfection
13.
J Drug Target ; 23(7-8): 736-49, 2015.
Article in English | MEDLINE | ID: mdl-26453169

ABSTRACT

Delivery of therapeutic agents to the central nervous system is a significant challenge, hindering progress in the treatment of diseases such as glioblastoma. Due to the presence of the blood-brain barrier (BBB), therapeutic agents do not readily transverse the brain endothelium to enter the parenchyma. Previous reports suggest that surface modification of polymer nanoparticles (NPs) can improve their ability to cross the BBB, but it is unclear whether the observed enhancements in transport are large enough to enhance therapy. In this study, we synthesized two degradable polymer NP systems surface-modified with ligands previously suggested to improve BBB transport, and tested their ability to cross the BBB after intravenous injection in mice. All the NP preparations were able to cross the BBB, although generally in low amounts (<0.5% of the injected dose), which was consistent with prior reports. One NP produced significantly higher brain uptake (∼0.8% of the injected dose): a block copolymer of polylactic acid and hyperbranched polyglycerol, surface modified with adenosine (PLA-HPG-Ad). PLA-HPG-Ad NPs provided controlled release of camptothecin, killing U87 glioma cells in culture. When administered intravenously in mice with intracranial U87 tumors, they failed to increase survival. These results suggest that enhancing NP transport across the BBB does not necessarily yield proportional pharmacological effects.


Subject(s)
Brain/metabolism , Camptothecin/administration & dosage , Drug Delivery Systems , Nanoparticles , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/pharmacology , Biological Transport , Blood-Brain Barrier/metabolism , Brain/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Camptothecin/pharmacokinetics , Camptothecin/pharmacology , Cell Line, Tumor , Delayed-Action Preparations , Drug Carriers/chemistry , Glioma/drug therapy , Glioma/pathology , Humans , Injections, Intravenous , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Polymers/chemistry , Tissue Distribution
14.
J Neurosci ; 35(26): 9764-81, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26134658

ABSTRACT

Peripheral and CNS inflammation leads to aberrations in developmental and postnatal neurogenesis, yet little is known about the mechanism linking inflammation to neurogenic abnormalities. Specific miRs regulate peripheral and CNS inflammatory responses. miR-155 is the most significantly upregulated miR in primary murine microglia stimulated with lipopolysaccharide (LPS), a proinflammatory Toll-Like Receptor 4 ligand. Here, we demonstrate that miR-155 is essential for robust IL6 gene induction in microglia under LPS stimulation in vitro. LPS-stimulated microglia enhance astrogliogenesis of cocultured neural stem cells (NSCs), whereas blockade of IL6 or genetic ablation of microglial miR-155 restores neural differentiation. miR-155 knock-out mice show reversal of LPS-induced neurogenic deficits and microglial activation in vivo. Moreover, mice with transgenic elevated expression of miR-155 in nestin-positive neural and hematopoietic stem cells, including microglia, show increased cell proliferation and ectopically localized doublecortin-positive immature neurons and radial glia-like cells in the hippocampal dentate gyrus (DG) granular cell layer. Microglia have proliferative and neurogenic effects on NSCs, which are significantly altered by microglial miR-155 overexpression. In addition, miR-155 elevation leads to increased microglial numbers and amoeboid morphology in the DG. Our study demonstrates that miR-155 is essential for inflammation-induced neurogenic deficits via microglial activation and induction of IL6 and is sufficient for disrupting normal hippocampal development.


Subject(s)
Gene Expression Regulation/genetics , Hippocampus/pathology , Inflammation/genetics , Inflammation/pathology , MicroRNAs/metabolism , Neurogenesis/genetics , Animals , Calcium-Binding Proteins/metabolism , Cell Culture Techniques , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Coculture Techniques , Disease Models, Animal , Doxycycline/pharmacology , Embryo, Mammalian , Female , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/drug effects , Inflammation/chemically induced , Interleukin-6/genetics , Interleukin-6/immunology , Interleukin-6/metabolism , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , Microfilament Proteins/metabolism , Nestin/genetics , Nestin/metabolism , Neurogenesis/drug effects , Pregnancy
15.
Nat Rev Drug Discov ; 14(4): 239-47, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25598505

ABSTRACT

The primary goal of nanomedicine is to improve clinical outcomes. To this end, targeted nanoparticles are engineered to reduce non-productive distribution while improving diagnostic and therapeutic efficacy. Paradoxically, as this field has matured, the notion of targeting has been minimized to the concept of increasing the affinity of a nanoparticle for its target. This Opinion article outlines a holistic view of nanoparticle targeting, in which the route of administration, molecular characteristics and temporal control of the nanoparticles are potential design variables that must be considered simultaneously. This comprehensive vision for nanoparticle targeting will facilitate the integration of nanomedicines into clinical practice.


Subject(s)
Drug Delivery Systems/methods , Nanoparticles/chemistry , Polymers/chemistry , Animals , Humans , Nanomedicine
16.
Nature ; 518(7537): 107-10, 2015 Feb 05.
Article in English | MEDLINE | ID: mdl-25409146

ABSTRACT

MicroRNAs are short non-coding RNAs expressed in different tissue and cell types that suppress the expression of target genes. As such, microRNAs are critical cogs in numerous biological processes, and dysregulated microRNA expression is correlated with many human diseases. Certain microRNAs, called oncomiRs, play a causal role in the onset and maintenance of cancer when overexpressed. Tumours that depend on these microRNAs are said to display oncomiR addiction. Some of the most effective anticancer therapies target oncogenes such as EGFR and HER2; similarly, inhibition of oncomiRs using antisense oligomers (that is, antimiRs) is an evolving therapeutic strategy. However, the in vivo efficacy of current antimiR technologies is hindered by physiological and cellular barriers to delivery into targeted cells. Here we introduce a novel antimiR delivery platform that targets the acidic tumour microenvironment, evades systemic clearance by the liver, and facilitates cell entry via a non-endocytic pathway. We find that the attachment of peptide nucleic acid antimiRs to a peptide with a low pH-induced transmembrane structure (pHLIP) produces a novel construct that could target the tumour microenvironment, transport antimiRs across plasma membranes under acidic conditions such as those found in solid tumours (pH approximately 6), and effectively inhibit the miR-155 oncomiR in a mouse model of lymphoma. This study introduces a new model for using antimiRs as anti-cancer drugs, which can have broad impacts on the field of targeted drug delivery.


Subject(s)
Drug Delivery Systems , Gene Expression Regulation, Neoplastic , Gene Silencing , Lymphoma/genetics , Lymphoma/therapy , MicroRNAs/antagonists & inhibitors , Tumor Microenvironment , Acids , Animals , Cell Membrane/metabolism , Cell Membrane Permeability , Disease Models, Animal , Female , Hydrogen-Ion Concentration , Lymphoma/pathology , Male , Mice , MicroRNAs/genetics , Molecular Targeted Therapy , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Oncogenes/genetics , Peptide Nucleic Acids/administration & dosage , Peptide Nucleic Acids/chemistry , Peptide Nucleic Acids/therapeutic use , Tumor Microenvironment/genetics
17.
Proc Natl Acad Sci U S A ; 111(30): E3062-71, 2014 Jul 29.
Article in English | MEDLINE | ID: mdl-25024194

ABSTRACT

Tumor suppressor p53 plays an important role in mediating growth inhibition upon telomere dysfunction. Here, we show that loss of the p53 target gene cyclin-dependent kinase inhibitor 1A (CDKN1A, also known as p21(WAF1/CIP1)) increases apoptosis induction following telomerase inhibition in a variety of cancer cell lines and mouse xenografts. This effect is highly specific to p21, as loss of other checkpoint proteins and CDK inhibitors did not affect apoptosis. In telomerase, inhibited cell loss of p21 leads to E2F1- and p53-mediated transcriptional activation of p53-upregulated modulator of apoptosis, resulting in increased apoptosis. Combined genetic or pharmacological inhibition of telomerase and p21 synergistically suppresses tumor growth. Furthermore, we demonstrate that simultaneous inhibition of telomerase and p21 also suppresses growth of tumors containing mutant p53 following pharmacological restoration of p53 activity. Collectively, our results establish that inactivation of p21 leads to increased apoptosis upon telomerase inhibition and thus identify a genetic vulnerability that can be exploited to treat many human cancers containing either wild-type or mutant p53.


Subject(s)
Apoptosis , Cyclin-Dependent Kinase Inhibitor p21/antagonists & inhibitors , Neoplasms, Experimental/metabolism , Telomerase/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Humans , Mice , Mice, Nude , Mutation , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Telomerase/genetics , Telomerase/metabolism , Tumor Suppressor Protein p53/genetics
18.
FASEB J ; 28(2): 908-22, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24221087

ABSTRACT

Transplantation of endothelial cells (ECs) for therapeutic vascularization or tissue engineering is a promising method for increasing tissue perfusion. Here, we report on a new approach for enhanced EC transplantation using targeted nanoparticle transfection to deliver proangiogenic microRNA-132 (miR-132) to cultured ECs before their transplantation, thereby sensitizing cells to the effects of endogenous growth factors. We synthesized biodegradable PLGA polymer nanoparticles (NPs) that were loaded with miR-132 and coated with cyclic RGD (cRGD) peptides that target integrin αvß3 expressed on cultured human umbilical vein ECs (HUVECs), increasing NP uptake through clathrin-coated pits. Unlike previously reported NPs for miR delivery, these NPs slowly release RNA for several weeks. The endocytosed NPs remain in clathrin-coated vesicles from which they mediate intracellular delivery of siRNA or miRNA. Transfection of HUVECs with miR-132 enhances growth factor-induced proliferation and migration in 2D culture, producing a 1.8- and 5-fold increase, respectively. However, while the effects of conventional transfection were short-lived, NP transfection produced protein knockdown and biological effects that were significantly longer in duration (≥ 6 d). Transfection of HUVECs with miR-132 NP resulted in a 2-fold increase in the number of microvessels per square millimeter compared to lipid after transplantation into immunodeficient mice and led to a higher number of mural cell-invested vessels than control transfection. These data suggest that sustained delivery of miR-132 encapsulated in a targeted biodegradable polymer NP is a safe and efficient strategy to improve EC transplantation and vascularization.


Subject(s)
MicroRNAs/administration & dosage , MicroRNAs/genetics , Nanoparticles/administration & dosage , Animals , Blotting, Western , Female , Flow Cytometry , Human Umbilical Vein Endothelial Cells/transplantation , Humans , Mice , Mice, SCID , Microscopy, Confocal , Nanotechnology/methods , Neovascularization, Physiologic , Reverse Transcriptase Polymerase Chain Reaction , Tissue Engineering , Transfection/methods
19.
Hear Res ; 304: 91-110, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23850665

ABSTRACT

The Notch pathway is a cell signaling pathway determining initial specification and subsequent cell fate in the inner ear. Previous studies have suggested that new hair cells (HCs) can be regenerated in the inner ear by manipulating the Notch pathway. In the present study, delivery of siRNA to Hes1 and Hes5 using a transfection reagent or siRNA to Hes1 encapsulated within poly(lactide-co-glycolide acid) (PLGA) nanoparticles increased HC numbers in non-toxin treated organotypic cultures of cochleae and maculae of postnatal day 3 mouse pups. An increase in HCs was also observed in cultured cochleae and maculae of mouse pups pre-conditioned with a HC toxin (4-hydroxy-2-nonenal or neomycin) and then treated with the various siRNA formulations. Treating cochleae with siRNA to Hes1 associated with a transfection reagent or siRNA to Hes1 delivered by PLGA nanoparticles decreased Hes1 mRNA and up-regulated Atoh1 mRNA expression allowing supporting cells (SCs) to acquire a HC fate. Experiments using cochleae and maculae of p27(kip1)/-GFP transgenic mouse pups demonstrated that newly generated HCs trans-differentiated from SCs. Furthermore, PLGA nanoparticles are non-toxic to inner ear tissue, readily taken up by cells within the tissue of interest, and present a synthetic delivery system that is a safe alternative to viral vectors. These results indicate that when delivered using a suitable vehicle, Hes siRNAs are potential therapeutic molecules that may have the capacity to regenerate new HCs in the inner ear and possibly restore human hearing and balance function.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Hair Cells, Auditory/physiology , Hair Cells, Vestibular/physiology , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Regeneration/genetics , Regeneration/physiology , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Aldehydes/toxicity , Animals , Hair Cells, Auditory/drug effects , Hair Cells, Vestibular/drug effects , Humans , Lactic Acid , Mice , Mice, Transgenic , Nanoparticles , Neomycin/toxicity , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Receptors, Notch/physiology , Tissue Culture Techniques , Transcription Factor HES-1
20.
Nanomedicine ; 9(8): 1124-34, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23764660

ABSTRACT

Nearly 40% of patients with non-invasive bladder cancer will progress to invasive disease despite locally-directed therapy. Overcoming the bladder permeability barrier (BPB) is a challenge for intravesical drug delivery. Using the fluorophore coumarin (C6), we synthesized C6-loaded poly(lactide-co-glycolide) (PLGA) nanoparticles (NPs), which were surface modified with a novel cell penetrating polymer, poly(guanidinium oxanorbornene) (PGON). Addition of PGON to the NP surface improved tissue penetration by 10-fold in intravesically-treated mouse bladder and ex vivo human ureter. In addition, NP-C6-PGON significantly enhanced intracellular uptake of NPs compared to NPs without PGON. To examine biological activity, we synthesized NPs that were loaded with the histone deacetylase (HDAC) inhibitor belinostat (NP-Bel-PGON). NP-Bel-PGON exhibited a significantly lower IC50 in cultured bladder cancer cells, and sustained hyperacetylation, when compared to unencapsulated belinostat. Xenograft tumors treated with NP-Bel-PGON showed a 70% reduction in volume, and a 2.5-fold higher intratumoral acetyl-H4, when compared to tumors treated with unloaded NP-PGON. FROM THE CLINICAL EDITOR: These authors demonstrate that PLGA nanoparticles with PGON surface functionalization result in greatly enhanced cell penetrating capabilities, and present convincing data from a mouse model of bladder cancer for increased chemotherapy efficacy.


Subject(s)
Drug Carriers/chemistry , Histone Deacetylase Inhibitors/administration & dosage , Hydroxamic Acids/administration & dosage , Nanoparticles/chemistry , Sulfonamides/administration & dosage , Urinary Bladder Neoplasms/drug therapy , Animals , Cell Line, Tumor , Drug Carriers/metabolism , Drug Delivery Systems , Female , Histone Deacetylase Inhibitors/pharmacokinetics , Histone Deacetylase Inhibitors/therapeutic use , Humans , Hydroxamic Acids/pharmacokinetics , Hydroxamic Acids/therapeutic use , Mice , Nanoparticles/metabolism , Polyglactin 910/chemistry , Polyglactin 910/metabolism , Sulfonamides/pharmacokinetics , Sulfonamides/therapeutic use , Urinary Bladder/drug effects , Urinary Bladder/metabolism , Urinary Bladder/pathology , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Urothelium/drug effects , Urothelium/metabolism , Urothelium/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...