Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 157
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 592(7853): 195-204, 2021 04.
Article in English | MEDLINE | ID: mdl-33828315

ABSTRACT

The move from reading to writing the human genome offers new opportunities to improve human health. The United States National Institutes of Health (NIH) Somatic Cell Genome Editing (SCGE) Consortium aims to accelerate the development of safer and more-effective methods to edit the genomes of disease-relevant somatic cells in patients, even in tissues that are difficult to reach. Here we discuss the consortium's plans to develop and benchmark approaches to induce and measure genome modifications, and to define downstream functional consequences of genome editing within human cells. Central to this effort is a rigorous and innovative approach that requires validation of the technology through third-party testing in small and large animals. New genome editors, delivery technologies and methods for tracking edited cells in vivo, as well as newly developed animal models and human biological systems, will be assembled-along with validated datasets-into an SCGE Toolkit, which will be disseminated widely to the biomedical research community. We visualize this toolkit-and the knowledge generated by its applications-as a means to accelerate the clinical development of new therapies for a wide range of conditions.


Subject(s)
Cells/metabolism , Gene Editing/methods , Genome, Human/genetics , National Institutes of Health (U.S.)/organization & administration , Animals , Genetic Therapy , Goals , Humans , United States
2.
Nature ; 582(7813): 586-591, 2020 06.
Article in English | MEDLINE | ID: mdl-32494005

ABSTRACT

Deregulation of metabolism and disruption of genome integrity are hallmarks of cancer1. Increased levels of the metabolites 2-hydroxyglutarate, succinate and fumarate occur in human malignancies owing to somatic mutations in the isocitrate dehydrogenase-1 or -2 (IDH1 or IDH2) genes, or germline mutations in the fumarate hydratase (FH) and succinate dehydrogenase genes (SDHA, SDHB, SDHC and SDHD), respectively2-4. Recent work has made an unexpected connection between these metabolites and DNA repair by showing that they suppress the pathway of homology-dependent repair (HDR)5,6 and confer an exquisite sensitivity to inhibitors of poly (ADP-ribose) polymerase (PARP) that are being tested in clinical trials. However, the mechanism by which these oncometabolites inhibit HDR remains poorly understood. Here we determine the pathway by which these metabolites disrupt DNA repair. We show that oncometabolite-induced inhibition of the lysine demethylase KDM4B results in aberrant hypermethylation of histone 3 lysine 9 (H3K9) at loci surrounding DNA breaks, masking a local H3K9 trimethylation signal that is essential for the proper execution of HDR. Consequently, recruitment of TIP60 and ATM, two key proximal HDR factors, is substantially impaired at DNA breaks, with reduced end resection and diminished recruitment of downstream repair factors. These findings provide a mechanistic basis for oncometabolite-induced HDR suppression and may guide effective strategies to exploit these defects for therapeutic gain.


Subject(s)
Chromatin/metabolism , DNA Repair , Homologous Recombination , Neoplasms/metabolism , Signal Transduction , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , Chromatin/drug effects , DNA Breaks/drug effects , DNA Repair/drug effects , Homologous Recombination/drug effects , Humans , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Lysine Acetyltransferase 5/metabolism , Methylation/drug effects , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Signal Transduction/drug effects
3.
Semin Cancer Biol ; 98: 11-18, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38029867

ABSTRACT

Hypoxia is a hallmark feature of the tumor microenvironment which can promote mutagenesis and instability. This increase in mutational burden occurs as a result of the downregulation of DNA repair systems. Deficits in the DNA damage response can be exploited to induce cytotoxicity and treat advanced stage cancers. With the advent of precision medicine, agents such as Poly (ADP-ribose) polymerase (PARP) inhibitors have been used to achieve synthetic lethality in homology directed repair (HDR) deficient cancers. However, most cancers lack these predictive biomarkers. Treatment for the HDR proficient population represents an important unmet clinical need. There has been interest in the use of anti-angiogenic agents to promote tumor hypoxia and induce deficiency in a HDR proficient background. For example, the use of cediranib to inhibit PDGFR and downregulate enzymes of the HDR pathway can be used synergistically with a PARP inhibitor. This combination can improve therapeutic responses in HDR proficient cancers. Preclinical results and Phase II and III clinical trial data support the mechanistic rationale for the efficacy of these agents in combination. Future investigations should explore the effectiveness of cediranib and other anti-angiogenic agents with a PARP inhibitor to elicit an antitumor response and sensitize cancers to immunotherapy.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Poly(ADP-ribose) Polymerases/metabolism , Poly(ADP-ribose) Polymerases/pharmacology , DNA Repair , Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Neoplasms/genetics , Hypoxia/genetics , Tumor Microenvironment
4.
Nucleic Acids Res ; 50(10): e59, 2022 06 10.
Article in English | MEDLINE | ID: mdl-35235944

ABSTRACT

Despite the rapid and broad implementation of CRISPR-Cas9-based technologies, convenient tools to modulate dose, timing, and precision remain limited. Building on methods using synthetic peptide nucleic acids (PNAs) to bind RNA with unusually high affinity, we describe guide RNA (gRNA) spacer-targeted, or 'antispacer', PNAs as a tool to modulate Cas9 binding and activity in cells in a sequence-specific manner. We demonstrate that PNAs rapidly and efficiently target complexed gRNA spacer sequences at low doses and without design restriction for sequence-selective Cas9 inhibition. We further show that short PAM-proximal antispacer PNAs achieve potent cleavage inhibition (over 2000-fold reduction) and that PAM-distal PNAs modify gRNA affinity to promote on-target specificity. Finally, we apply antispacer PNAs for temporal regulation of two dCas9-fusion systems. These results present a novel rational approach to nucleoprotein engineering and describe a rapidly implementable antisense platform for CRISPR-Cas9 modulation to improve spatiotemporal versatility and safety across applications.


Subject(s)
Peptide Nucleic Acids , RNA, Guide, Kinetoplastida , CRISPR-Cas Systems , Gene Editing/methods , Peptide Nucleic Acids/pharmacology , RNA, Guide, Kinetoplastida/genetics
5.
J Biol Chem ; 298(10): 102398, 2022 10.
Article in English | MEDLINE | ID: mdl-35988651

ABSTRACT

Unusual nucleic acid structures play vital roles as intermediates in many cellular processes and, in the case of peptide nucleic acid (PNA)-mediated triplexes, are leveraged as tools for therapeutic gene editing. However, due to their transient nature, an understanding of the factors that interact with and process dynamic nucleic acid structures remains limited. Here, we developed snapELISA (structure-specific nucleic acid-binding protein ELISA), a rapid high-throughput platform to interrogate and compare up to 2688 parallel nucleic acid structure-protein interactions inĀ vitro. We applied this system to both triplex-forming oligonucleotide-induced DNA triplexes and DNA-bound PNA heterotriplexes to describe the identification of previously known and novel interactors for both structures. For PNA heterotriplex recognition analyses, snapELISA identified factors implicated in nucleotide excision repair (XPA, XPC), single-strand annealing repair (RAD52), and recombination intermediate structure binding (TOP3A, BLM, MUS81). We went on to validate selected factor localization to genome-targeted PNA structures within clinically relevant loci in human cells. Surprisingly, these results demonstrated XRCC5 localization to PNA triplex-forming sites in the genome, suggesting the presence of a double-strand break intermediate. These results describe a powerful comparative approach for identifying structure-specific nucleic acid interactions and expand our understanding of the mechanisms of triplex structure recognition and repair.


Subject(s)
DNA , Peptide Nucleic Acids , Humans , DNA/chemistry , DNA/metabolism , Enzyme-Linked Immunosorbent Assay , Nucleic Acid Conformation , Peptide Nucleic Acids/chemistry , Peptide Nucleic Acids/metabolism
6.
Mol Cell ; 55(1): 31-46, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24882211

ABSTRACT

MutS protein homolog 2 (MSH2) is a key DNA mismatch repair protein. It forms the MSH2-MSH6 (MutSα) and MSH2-MSH3 (MutSƟ) heterodimers, which help to ensure genomic integrity. MutSα not only recognizes and repairs mismatched nucleotides but also recognizes DNA adducts induced by DNA-damaging agents, and triggers cell-cycle arrest and apoptosis. Loss or depletion of MutSα from cells leads to microsatellite instability (MSI) and resistance to DNA damage. Although the level of MutSα can be reduced by the ubiquitin-proteasome pathway, the detailed mechanisms of this regulation remain elusive. Here we report that histone deacetylase 6 (HDAC6) sequentially deacetylates and ubiquitinates MSH2, leading to MSH2 degradation. In addition, HDAC6 significantly reduces cellular sensitivity to DNA-damaging agents and decreases cellular DNA mismatch repair activities by downregulation of MSH2. Overall, these findings reveal a mechanism by which proper levels of MutSα are maintained.


Subject(s)
Histone Deacetylases/physiology , MutS Homolog 2 Protein/metabolism , Acetylation , Animals , Cells, Cultured , HEK293 Cells , HeLa Cells , Histone Deacetylase 6 , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Mice , Protein Stability , Ubiquitination
7.
Oncologist ; 26(7): e1104-e1109, 2021 07.
Article in English | MEDLINE | ID: mdl-33742489

ABSTRACT

LESSONS LEARNED: Cediranib and olaparib combination did not result in clinically meaningful activity in patients with metastatic pancreatic ductal adenocarcinoma without known BRCA mutation. BACKGROUND: Cediranib, a vascular endothelial growth factor receptor inhibitor, suppresses expression of BRCA1/2 and RAD51 inducing homologous recombination DNA repair deficiency (HRD) in several cancer cell lines and xenograft models [1]. Olaparib provides a clinical benefit in patients with metastatic pancreatic adenocarcinoma (mPDAC) with germline BRCA mutation (gBRCAmt) [2]. We hypothesized that cediranib induces HRD in the absence of gBRCAmt and synergizes with olaparib, resulting in an objective response in patients with mPDAC. METHODS: Patients with mPDAC with at least one prior systemic chemotherapy were enrolled. Patients with known gBRCAmt were excluded. Patients took cediranib 30 mg daily and olaparib 200 mg twice daily, orally. The primary endpoint was objective response (OR) rate. RESULTS: Nineteen patients received the study drugs. Seven patients came off treatment before the first restaging scan: six because of clinical progression and one because of an adverse event. No OR was observed. Six patients had stable disease (SD) as a best overall response. The median duration of SD was 3.1 months. The median overall survival was 3.4 months. Common treatment-related adverse events were fatigue, hypertension, and diarrhea. CONCLUSION: Cediranib and olaparib combination did not result in clinically meaningful activity in patients with mPDAC without gBRCAmt.


Subject(s)
Adenocarcinoma , Ovarian Neoplasms , Pancreatic Neoplasms , Female , Humans , Mutation , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Phthalazines/adverse effects , Piperazines , Quinazolines , Vascular Endothelial Growth Factor A
8.
Biopolymers ; 112(12): e23460, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34129732

ABSTRACT

The unique properties of peptide nucleic acid (PNA) makes it a desirable candidate to be used in therapeutic and biotechnological interventions. It has been broadly utilized for numerous applications, with a major focus in regulation of gene expression, and more recently in gene editing. While the classic PNA design has mainly been employed to date, chemical modifications of the PNA backbone and nucleobases provide an avenue to advance the technology further. This review aims to discuss the recent developments in PNA based gene manipulation techniques and the use of novel chemical modifications to improve the current state of PNA mediated gene targeting.


Subject(s)
Peptide Nucleic Acids , Gene Expression Regulation
9.
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
10.
Int J Mol Sci ; 22(23)2021 Nov 25.
Article in English | MEDLINE | ID: mdl-34884568

ABSTRACT

Maintenance of genomic integrity is crucial for cell survival. As such, elegant DNA damage response (DDR) systems have evolved to ensure proper repair of DNA double-strand breaks (DSBs) and other lesions that threaten genomic integrity. Towards this end, most therapeutic studies have focused on understanding of the canonical DNA DSB repair pathways to enhance the efficacy of DNA-damaging therapies. While these approaches have been fruitful, there has been relatively limited success to date and potential for significant normal tissue toxicity. With the advent of novel immunotherapies, there has been interest in understanding the interactions of radiation therapy with the innate and adaptive immune responses, with the ultimate goal of enhancing treatment efficacy. While a substantial body of work has demonstrated control of the immune-mediated (extrinsic) responses to DNA-damaging therapies by several innate immune pathways (e.g., cGAS-STING and RIG-I), emerging work demonstrates an underappreciated role of the innate immune machinery in directly regulating tumor cell-intrinsic/cell-autonomous responses to DNA damage.


Subject(s)
DNA Damage , DNA Repair , Immunity, Innate , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , Humans , Membrane Proteins/genetics , Nucleotidyltransferases/genetics , Signal Transduction
11.
Mutagenesis ; 35(1): 61-68, 2020 02 13.
Article in English | MEDLINE | ID: mdl-31282537

ABSTRACT

Hypoxia is a hallmark of the tumour microenvironment with profound effects on tumour biology, influencing cancer progression, the development of metastasis and patient outcome. Hypoxia also contributes to genomic instability and mutation frequency by inhibiting DNA repair pathways. This review summarises the diverse mechanisms by which hypoxia affects DNA repair, including suppression of homology-directed repair, mismatch repair and base excision repair. We also discuss the effects of hypoxia mimetics and agents that induce hypoxia on DNA repair, and we highlight areas of potential clinical relevance as well as future directions.


Subject(s)
DNA Repair , Neoplasms/genetics , Neoplasms/metabolism , Cell Hypoxia , DNA Mismatch Repair , Gene Expression Regulation, Neoplastic , Genome , Genomic Instability , Humans , Molecular Targeted Therapy , Neoplasm Metastasis , Neoplasms/pathology , Recombinational DNA Repair , Tumor Hypoxia/drug effects , Tumor Microenvironment/genetics
12.
Mol Pharm ; 17(2): 461-471, 2020 02 03.
Article in English | MEDLINE | ID: mdl-31855437

ABSTRACT

Tumor-targeted drug delivery systems offer not only the advantage of an enhanced therapeutic index, but also the possibility of overcoming the limitations that have largely restricted drug design to small, hydrophobic, "drug-like" molecules. Here, we explore the ability of a tumor-targeted delivery system centered on the use of aĀ pH-low insertion peptide (pHLIP) to directly deliver moderately polar, multi-kDa molecules into tumor cells. A pHLIP is a short, pH-responsive peptide capable of inserting across a cell membrane to form a transmembrane helix at acidic pH. pHLIPs target the acidic tumor microenvironment with high specificity, and a drug attached to the inserting end of a pHLIP can be translocated across the cell membrane during the insertion process. We investigate the ability of wildtypeĀ pHLIP to deliver peptide nucleic acid (PNA) cargoes of varying sizes across lipid membranes. We find that pHLIP effectively delivers PNAs up to Ć¢ĀˆĀ¼7 kDa into cells in a pH-dependent manner. In addition, pHLIP retainsĀ its tumor-targeting capabilities when linked to cargoes of this size, although the amount delivered is reduced for PNA cargoes greater than Ć¢ĀˆĀ¼6 kDa. As drug-like molecules are traditionally restricted to sizes of Ć¢ĀˆĀ¼500 Da, this constitutes an order-of-magnitude expansion in the size range of deliverable drug candidates.


Subject(s)
Cytoplasm/drug effects , Drug Delivery Systems/methods , Melanoma/drug therapy , Membrane Proteins/metabolism , Peptide Nucleic Acids/administration & dosage , Skin Neoplasms/drug therapy , A549 Cells , Animals , Cell Membrane/metabolism , Cell Membrane Permeability/drug effects , Disease Models, Animal , Humans , Hydrogen-Ion Concentration , Lipid Bilayers/metabolism , Melanoma/pathology , Membrane Proteins/pharmacology , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy/methods , Skin Neoplasms/pathology , Treatment Outcome , Tumor Microenvironment/drug effects
13.
Molecules ; 25(3)2020 Feb 08.
Article in English | MEDLINE | ID: mdl-32046275

ABSTRACT

Unusual nucleic acid structures are salient triggers of endogenous repair and can occur in sequence-specific contexts. Peptide nucleic acids (PNAs) rely on these principles to achieve non-enzymatic gene editing. By forming high-affinity heterotriplex structures within the genome, PNAs have been used to correct multiple human disease-relevant mutations with low off-target effects. Advances in molecular design, chemical modification, and delivery have enabled systemic in vivo application of PNAs resulting in detectable editing in preclinical mouse models. In a model of Ɵ-thalassemia, treated animals demonstrated clinically relevant protein restoration and disease phenotype amelioration, suggesting a potential for curative therapeutic application of PNAs to monogenic disorders. This review discusses the rationale and advances of PNA technologies and their application to gene editing with an emphasis on structural biochemistry and repair.


Subject(s)
Cystic Fibrosis/therapy , DNA/genetics , Gene Editing/methods , Genetic Therapy/methods , Peptide Nucleic Acids/genetics , beta-Thalassemia/therapy , Animals , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis/pathology , DNA/metabolism , Disease Models, Animal , Gene Targeting/methods , Gene Transfer Techniques , Humans , Mice , Nanoparticles/chemistry , Nanoparticles/metabolism , Nucleic Acid Conformation , Nucleic Acid Hybridization , Peptide Nucleic Acids/administration & dosage , Peptide Nucleic Acids/metabolism , Recombinational DNA Repair , beta-Thalassemia/genetics , beta-Thalassemia/metabolism , beta-Thalassemia/pathology
14.
Nucleic Acids Res ; 45(20): 11782-11799, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29036688

ABSTRACT

RAD51, a key factor in homology-directed repair (HDR), has long been considered an attractive target for cancer therapy, but few specific inhibitors have been found. A cell-penetrating, anti-DNA, lupus autoantibody, 3E10, was previously shown to inhibit HDR, sensitize tumors to radiation, and mediate synthetic lethal killing of BRCA2-deficient cancer cells, effects that were initially attributed to its affinity for DNA. However, as the molecular basis for its ability to inhibit DNA repair, we report that 3E10 directly binds to the N-terminus of RAD51, sequesters RAD51 in the cytoplasm, and impedes RAD51 binding to DNA. Further, we generate separation-of-function mutations in the complementarity-determining regions of 3E10 revealing that inhibition of HDR tracks with binding to RAD51 but not to DNA, whereas cell penetration is linked to DNA binding. The consequences of these mutations on putative 3E10 interactions with RAD51 and DNA are correlated with in silico molecular modeling. Taken together, the results identify 3E10 as a novel inhibitor of RAD51 by direct binding, accounting for its ability to suppress HDR and providing the molecular basis to guide pre-clinical development of 3E10 as an anti-cancer agent.


Subject(s)
Autoantibodies/metabolism , DNA Repair , DNA/metabolism , Rad51 Recombinase/metabolism , Autoantibodies/chemistry , Autoantibodies/genetics , Cell Line, Tumor , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/genetics , Cell-Penetrating Peptides/metabolism , Cells, Cultured , Complementarity Determining Regions/genetics , Cytoplasm/metabolism , DNA/chemistry , DNA/genetics , HEK293 Cells , Humans , Lupus Erythematosus, Systemic/immunology , Models, Molecular , Mutation , Protein Binding , Protein Domains , Rad51 Recombinase/chemistry , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics , Single-Chain Antibodies/metabolism
15.
Breast Cancer Res ; 20(1): 16, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29510720

ABSTRACT

BACKGROUND: Tumor hypoxia is an independent prognostic factor associated with poor patient survival. Emerging evidence suggests that hypoxia can potentially maintain or enhance the stem cell phenotype of both normal stem cells and cancer cells. However, it remains to be determined whether cell fate is regulated in vivo by the hypoxic tumor microenvironment (TME). METHODS: We established a hypoxia-sensing xenograft model to identify hypoxic tumor cell in vivo primarily using human breast cancer cell lines MDA-MB-231 and MCF7. Hypoxic tumor cells were identified in situ by fluorescence of green fluorescence protein. They were further isolated from xenografts, purified and sorted by flow cytometry for detailed analysis of their stem cell characteristics. RESULTS: We have found that hypoxic tumor cells freshly isolated from xenografts contain increased subpopulations of tumor cells with cancer stem cell (CSC)-like characteristics. The CSC characteristics of the hypoxic tumor cells are further enhanced upon re-implantation in vivo, whereas secondary xenografts derived from the non-hypoxic tumor cells remain similar to the primary xenografts. Interestingly, the phenotypes exhibited by the hypoxic tumor cells are stable and remain distinctively different from those of the non-hypoxic tumor cells isolated from the same tumor mass even when they are maintained under the same ambient culture conditions. Mechanistically, the PI3K/AKT pathway is strongly potentiated in the hypoxic tumor cells and is required to maintain the CSC-like phenotype. Importantly, the differential cell fates between hypoxic and non-hypoxic tumor cells are only found in tumor cells isolated from the hypoxic TME in vivo and are not seen in tumor cells treated by hypoxia in vitro alone. CONCLUSIONS: These previously unknown observations suggest that the hypoxic TME may promote malignant progression and therapy resistance by coordinating induction, selection and/or preferential maintenance of the CSC-like phenotype in tumor cells.


Subject(s)
Breast Neoplasms/genetics , Neoplastic Stem Cells/metabolism , Tumor Hypoxia/genetics , Tumor Microenvironment/genetics , Animals , Breast Neoplasms/pathology , Cell Differentiation/genetics , Cell Lineage/genetics , Female , Humans , MCF-7 Cells , Mice , Neoplastic Stem Cells/pathology , Xenograft Model Antitumor Assays
17.
Molecules ; 23(3)2018 Mar 11.
Article in English | MEDLINE | ID: mdl-29534473

ABSTRACT

Peptide nucleic acids (PNAs) can bind duplex DNA in a sequence-targeted manner, forming a triplex structure capable of inducing DNA repair and producing specific genome modifications. Since the first description of PNA-mediated gene editing in cell free extracts, PNAs have been used to successfully correct human disease-causing mutations in cell culture and in vivo in preclinical mouse models. Gene correction via PNAs has resulted in clinically-relevant functional protein restoration and disease improvement, with low off-target genome effects, indicating a strong therapeutic potential for PNAs in the treatment or cure of genetic disorders. This review discusses the progress that has been made in developing PNAs as an effective, targeted agent for gene editing, with an emphasis on recent in vivo, nanoparticle-based strategies.


Subject(s)
DNA/metabolism , Gene Editing/methods , Peptide Nucleic Acids/pharmacology , Animals , DNA/chemistry , Disease Models, Animal , Genetic Predisposition to Disease , Humans , Mice , Mutagenesis, Site-Directed , Nucleic Acid Conformation , Peptide Nucleic Acids/chemistry , Peptide Nucleic Acids/therapeutic use
18.
Carcinogenesis ; 38(6): 627-637, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28472268

ABSTRACT

The heavy metal nickel is a known carcinogen, and occupational exposure to nickel compounds has been implicated in human lung and nasal cancers. Unlike many other environmental carcinogens, however, nickel does not directly induce DNA mutagenesis, and the mechanism of nickel-related carcinogenesis remains incompletely understood. Cellular nickel exposure leads to signaling pathway activation, transcriptional changes and epigenetic remodeling, processes also impacted by hypoxia, which itself promotes tumor growth without causing direct DNA damage. One of the mechanisms by which hypoxia contributes to tumor growth is the generation of genomic instability via down-regulation of high-fidelity DNA repair pathways. Here, we find that nickel exposure similarly leads to down-regulation of DNA repair proteins involved in homology-dependent DNA double-strand break repair (HDR) and mismatch repair (MMR) in tumorigenic and non-tumorigenic human lung cells. Functionally, nickel induces a defect in HDR capacity, as determined by plasmid-based host cell reactivation assays, persistence of ionizing radiation-induced DNA double-strand breaks and cellular hypersensitivity to ionizing radiation. Mechanistically, we find that nickel, in contrast to the metalloid arsenic, acutely induces transcriptional repression of HDR and MMR genes as part of a global transcriptional pattern similar to that seen with hypoxia. Finally, we find that exposure to low-dose nickel reduces the activity of the MLH1 promoter, but only arsenic leads to long-term MLH1 promoter silencing. Together, our data elucidate novel mechanisms of heavy metal carcinogenesis and contribute to our understanding of the influence of the microenvironment on the regulation of DNA repair pathways.


Subject(s)
DNA Repair Enzymes , DNA Repair/drug effects , Lung Neoplasms/genetics , Lung/metabolism , Nickel/toxicity , Trace Elements/toxicity , Arsenites/toxicity , Cell Hypoxia/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/genetics , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/radiation effects , Down-Regulation , Genomic Instability , Humans , Lung/pathology , Lung Neoplasms/pathology , Teratogens/toxicity
19.
Nucleic Acids Res ; 43(1): 259-71, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25488815

ABSTRACT

Serine/threonine kinase 11 (STK11, also known as LKB1) functions as a tumor suppressor in many human cancers. However, paradoxically loss of LKB1 in mouse embryonic fibroblast results in resistance to oncogene-induced transformation. Therefore, it is unclear why loss of LKB1 leads to increased predisposition to develop a wide variety of cancers. Here, we show that LKB1 protects cells from genotoxic stress. Cells lacking LKB1 display increased sensitivity to irradiation, accumulates more DNA double-strand breaks, display defective homology-directed DNA repair (HDR) and exhibit increased mutation rate, compared with that of LKB1-expressing cells. Conversely, the ectopic expression of LKB1 in cells lacking LKB1 protects them against genotoxic stress-induced DNA damage and prevents the accumulation of mutations. We find that LKB1 post-transcriptionally stimulates HDR gene BRCA1 expression by inhibiting the cytoplasmic localization of the RNA-binding protein, HU antigen R, in an AMP kinase-dependent manner and stabilizes BRCA1 mRNA. Cells lacking BRCA1 similar to the cell lacking LKB1 display increased genomic instability and ectopic expression of BRCA1 rescues LKB1 loss-induced sensitivity to genotoxic stress. Collectively, our results demonstrate that LKB1 is a crucial regulator of genome integrity and reveal a novel mechanism for LKB1-mediated tumor suppression with direct therapeutic implications for cancer prevention.


Subject(s)
BRCA1 Protein/genetics , Protein Serine-Threonine Kinases/physiology , Recombinational DNA Repair , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/metabolism , Animals , BRCA1 Protein/metabolism , Cell Cycle , Cell Line , DNA Breaks, Double-Stranded , DNA Damage , ELAV Proteins/metabolism , Genome , Humans , Mice , Mutation , RNA Stability , RNA, Messenger/metabolism
20.
Yale J Biol Med ; 90(4): 583-598, 2017 12.
Article in English | MEDLINE | ID: mdl-29259523

ABSTRACT

Since their invention in 1991, peptide nucleic acids (PNAs) have been used in a myriad of chemical and biological assays. More recently, peptide nucleic acids have also been demonstrated to hold great potential as therapeutic agents because of their physiological stability, affinity for target nucleic acids, and versatility. While recent modifications in their design have further improved their potency, their preclinical development has reached new heights due to their combination with recent advancements in drug delivery. This review focuses on recent advances in PNA therapeutic applications, in which chemical modifications are made to improve PNA function and nanoparticles are used to enhance PNA delivery.


Subject(s)
Drug Delivery Systems/methods , Gene Expression Regulation/drug effects , Peptide Nucleic Acids/administration & dosage , Peptide Nucleic Acids/therapeutic use , Antisense Elements (Genetics) , Drug Stability , Gene Editing , Humans , MicroRNAs , Molecular Structure , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Peptide Nucleic Acids/chemistry , Peptide Nucleic Acids/pharmacokinetics , Solubility
SELECTION OF CITATIONS
SEARCH DETAIL