Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 246
Filter
1.
Adv Drug Deliv Rev ; 175: 113809, 2021 08.
Article in English | MEDLINE | ID: mdl-34033819

ABSTRACT

Thanks to their abilities to modulate the expression of virtually any genes, RNA therapeutics have attracted considerable research efforts. Among the strategies focusing on nucleic acid gene inhibitors, antisense oligonucleotides and small interfering RNAs have reached advanced clinical trial phases with several of them having recently been marketed. These successes were obtained by overcoming stability and cellular delivery issues using either chemically modified nucleic acids or nanoparticles. As nucleic acid gene inhibitors are promising strategies to treat inflammatory diseases, this review focuses on the barriers, from manufacturing issues to cellular/subcellular delivery, that still need to be overcome to deliver the nucleic acids to sites of inflammation other than the liver. Furthermore, key examples of applications in rheumatoid arthritis, inflammatory bowel, and lung diseases are presented as case studies of systemic, oral, and lung nucleic acid delivery.


Subject(s)
Inflammation/drug therapy , Nanomedicine/methods , Nanoparticle Drug Delivery System , Nucleic Acids/administration & dosage , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/therapeutic use , Drug Delivery Systems/methods , Genes/drug effects , Humans , Inflammation/genetics , Nucleic Acids/therapeutic use , Oligodeoxyribonucleotides, Antisense/administration & dosage , Oligodeoxyribonucleotides, Antisense/therapeutic use , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/therapeutic use
2.
Eur J Pharm Biopharm ; 162: 92-98, 2021 May.
Article in English | MEDLINE | ID: mdl-33753212

ABSTRACT

This study describes the effective attack of oligonucleotides on the viral genome of highly pathogenic H5N1 influenza A virus (IAV) in vivo using for the first time the new delivery system consisting of biocompatible low-toxic titanium dioxide nanoparticles and immobilized polylysine-containing oligonucleotides with the native (ODN) and partially modified (ODNm) internucleotide bonds. Intraperitoneal injection of the TiO2•PL-ODN nanocomposite provided 65-70% survival of mice, while intraperitoneal or oral administration of TiO2•PL-ODNm was somewhat more efficient (~80% survival). The virus titer in the lung was reduced by two-three orders of magnitude. The nanocomposites are nontoxic to mice under the used conditions. TiO2 nanoparticles, unbound ODN, and the nanocomposite bearing the random oligonucleotide showed an insignificant protective effect, which indicates the ability of targeted oligonucleotides delivered in mice in the nanocomposites to site-specifically interact with complementary RNAs. The protection of oligonucleotides in nanocomposites by TiO2 nanoparticles and partial modification of the internucleotide bonds provides a continued presence of oligonucleotides in the body for the effective and specific action on the viral RNA. The proposed oligonucleotide delivery system can claim not only to effectively inhibit IAV genes but also to turn off other genes responsible for diseases caused by nucleic acids.


Subject(s)
Antiviral Agents/administration & dosage , Drug Carriers/chemistry , Influenza A Virus, H5N1 Subtype/drug effects , Influenza, Human/drug therapy , Oligodeoxyribonucleotides, Antisense/administration & dosage , Administration, Oral , Animals , Disease Models, Animal , Dogs , Female , Genome, Viral/drug effects , Humans , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/isolation & purification , Influenza, Human/virology , Injections, Intraperitoneal , Madin Darby Canine Kidney Cells , Male , Mice , Nanocomposites/chemistry , RNA, Viral/antagonists & inhibitors , Titanium/chemistry , Viral Load/drug effects
3.
Nucleic Acid Ther ; 30(2): 94-103, 2020 04.
Article in English | MEDLINE | ID: mdl-32043907

ABSTRACT

Inotersen, a 2'-O-methoxyethyl (2'-MOE) phosphorothioate antisense oligonucleotide, reduced disease progression and improved quality of life in patients with hereditary transthyretin amyloidosis with polyneuropathy (hATTR-PN) in the NEURO-TTR and NEURO-TTR open-label extension (OLE) trials. However, 300 mg/week inotersen treatment was associated with platelet count reductions in several patients. Mean platelet counts in patients in the NEURO-TTR-inotersen group remained ≥140 × 109/L in 50% and ≥100 × 109/L in 80% of the subjects. However, grade 4 thrombocytopenia (<25 × 109/L) occurred in three subjects in NEURO-TTR trial, and one of these suffered a fatal intracranial hemorrhage. The two others were treated successfully with corticosteroids and discontinuation of inotersen. Investigations in a subset of subjects in NEURO-TTR (n = 17 placebo; n = 31 inotersen) and OLE (n = 33) trials ruled out direct myelotoxicity, consumptive coagulopathy, and heparin-induced thrombocytopenia. Antiplatelet immunoglobulin G (IgG) antibodies were detected at baseline in 5 of 31 (16%) inotersen-treated subjects in NEURO-TTR, 4 of whom eventually developed grade 1 or 2 thrombocytopenia while on the drug. In addition, 24 subjects in the same group developed treatment-emergent antiplatelet IgG antibodies, of which 2 developed grade 2, and 3 developed grade 4 thrombocytopenia. Antiplatelet IgG antibodies in two of the three grade 4 thrombocytopenia subjects targeted GPIIb/IIIa. Plasma cytokines previously implicated in immune dysregulation, such as interleukin (IL)-23 and a proliferation-inducing ligand (APRIL) were often above the normal range at baseline. Collectively, these findings suggest an underlying immunologic dysregulation predisposing some individuals to immune-mediated thrombocytopenia during inotersen treatment.


Subject(s)
Amyloid Neuropathies, Familial/drug therapy , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides/administration & dosage , Thrombocytopenia/blood , Adult , Aged , Amyloid Neuropathies, Familial/genetics , Amyloid Neuropathies, Familial/immunology , Amyloid Neuropathies, Familial/pathology , Female , Genetic Predisposition to Disease , Humans , Immune System Diseases/chemically induced , Immune System Diseases/immunology , Immune System Diseases/pathology , Immunoglobulin G , Intracranial Hemorrhages/chemically induced , Intracranial Hemorrhages/immunology , Intracranial Hemorrhages/pathology , Male , Middle Aged , Oligodeoxyribonucleotides, Antisense/administration & dosage , Oligonucleotides/adverse effects , Oligonucleotides, Antisense/adverse effects , Quality of Life , Thrombocytopenia/chemically induced , Thrombocytopenia/immunology , Thrombocytopenia/pathology
4.
IUBMB Life ; 72(5): 855-871, 2020 05.
Article in English | MEDLINE | ID: mdl-31913572

ABSTRACT

Multidrug resistance (MDR), defined as the ability of cancer cells to gain resistance to both conventional and novel chemotherapy agents, is an important barrier in treating malignancies. Initially, it was discovered that cellular pumps dependent on ATP were the cause of resistance to chemotherapy, and further studies have found that other mechanisms such as increased metabolism of drugs, decreased drug entry, and defective apoptotic pathways are involved in this process. MDR has been the focus of numerous initiatives and countless studies have been undertaken to better understand MDR and formulate strategies to overcome its effects. The current review highlights various nano-drug delivery systems including polymeric/solid lipid/mesoporous silica/metal nanoparticles, dendrimers, liposomes, micelles, and nanostructured lipid carriers to overcome the mechanism of MDR. Nanoparticles are novel gateways to enhance the therapeutic efficacy of anticancer agents at the target site of action due to their tumor-targeting abilities, which can limit the unwanted systemic effects of chemotherapy agents and also reduce drug resistance. Additionally, other innovative strategies including RNA interference as a biological process used to inhibit or silence specific gene expression, natural products as MDR modulators with little systemic toxic effects, which interfere with the functions of proteins involved in drug efflux, and physical approaches such as combination of conventional drug administration with thermal/ultrasound/photodynamic strategies are also highlighted.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Carriers , Drug Resistance, Neoplasm/drug effects , Molecular Targeted Therapy/methods , Nanotechnology/methods , Neoplasms/therapy , Animals , Antineoplastic Agents/metabolism , Cell Line, Tumor , Dendrimers/chemistry , Dendrimers/pharmacokinetics , Drug Compounding/methods , Humans , Liposomes/chemistry , Liposomes/pharmacokinetics , Mice , Micelles , Nanoparticles/chemistry , Nanoparticles/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Oligodeoxyribonucleotides, Antisense/administration & dosage , Oligodeoxyribonucleotides, Antisense/genetics , Oligodeoxyribonucleotides, Antisense/metabolism , Xenograft Model Antitumor Assays
5.
J Neurol ; 267(4): 1070-1079, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31853709

ABSTRACT

OBJECTIVE: To examine the impact on quality of life (QOL) of patients with hATTR amyloidosis with polyneuropathy treated with inotersen (Tegsedi™) versus placebo. METHODS: Data were from the NEURO-TTR trial (ClinicalTrials.gov Identifier: NCT01737398), a phase 3, multinational, randomized, double-blind, placebo-controlled study of inotersen in patients with hATTR amyloidosis with polyneuropathy. At baseline and week 66, QOL measures-the Norfolk-QOL-Diabetic Neuropathy (DN) questionnaire and SF-36v2® Health Survey (SF-36v2)-were assessed. Treatment differences in mean changes in QOL from baseline to week 66 were tested using mixed-effect models with repeated measures. Responder analyses compared the percentages of patients whose QOL meaningfully improved or worsened from baseline to week 66 in inotersen and placebo arms. Descriptive analysis of item responses examined treatment differences in specific activities and functions at week 66. RESULTS: Statistically significant mean differences between treatment arms were observed for three of five Norfolk-QOL-DN domains and five of eight SF-36v2 domains, with better outcomes for inotersen than placebo in physical functioning, activities of daily living, neuropathic symptoms, pain, role limitations due to health problems, and social functioning. A larger percentage of patients in the inotersen arm than the placebo arm showed preservation or improvement in Norfolk-QOL-DN and SF-36v2 scores from baseline to week 66. Responses at week 66 showed more substantial problems with daily activities and functioning for patients in the placebo arm than in the inotersen arm. CONCLUSION: Patients with hATTR amyloidosis with polyneuropathy treated with inotersen showed preserved or improved QOL at 66 weeks compared to those who received placebo.


Subject(s)
Activities of Daily Living , Amyloid Neuropathies, Familial/drug therapy , Oligodeoxyribonucleotides, Antisense/pharmacology , Oligonucleotides/pharmacology , Outcome Assessment, Health Care , Polyneuropathies/drug therapy , Quality of Life , Severity of Illness Index , Adult , Aged , Aged, 80 and over , Amyloid Neuropathies, Familial/complications , Double-Blind Method , Drug Administration Schedule , Female , Humans , Male , Middle Aged , Mobility Limitation , Oligodeoxyribonucleotides, Antisense/administration & dosage , Oligonucleotides/administration & dosage , Polyneuropathies/etiology
6.
J Clin Invest ; 129(11): 4817-4831, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31589162

ABSTRACT

BACKGROUNDSpinal muscular atrophy (SMA) is caused by deficient expression of survival motor neuron (SMN) protein. New SMN-enhancing therapeutics are associated with variable clinical benefits. Limited knowledge of baseline and drug-induced SMN levels in disease-relevant tissues hinders efforts to optimize these treatments.METHODSSMN mRNA and protein levels were quantified in human tissues isolated during expedited autopsies.RESULTSSMN protein expression varied broadly among prenatal control spinal cord samples, but was restricted at relatively low levels in controls and SMA patients after 3 months of life. A 2.3-fold perinatal decrease in median SMN protein levels was not paralleled by comparable changes in SMN mRNA. In tissues isolated from nusinersen-treated SMA patients, antisense oligonucleotide (ASO) concentration and full-length (exon 7 including) SMN2 (SMN2-FL) mRNA level increases were highest in lumbar and thoracic spinal cord. An increased number of cells showed SMN immunolabeling in spinal cord of treated patients, but was not associated with an increase in whole-tissue SMN protein levels.CONCLUSIONSA normally occurring perinatal decrease in whole-tissue SMN protein levels supports efforts to initiate SMN-inducing therapies as soon after birth as possible. Limited ASO distribution to rostral spinal and brain regions in some patients likely limits clinical response of motor units in these regions for those patients. These results have important implications for optimizing treatment of SMA patients and warrant further investigations to enhance bioavailability of intrathecally administered ASOs.FUNDINGSMA Foundation, SMART, NIH (R01-NS096770, R01-NS062869), Ionis Pharmaceuticals, and PTC Therapeutics. Biogen provided support for absolute real-time RT-PCR.


Subject(s)
Aging , Motor Neurons , Muscular Atrophy, Spinal , Oligodeoxyribonucleotides, Antisense/administration & dosage , Spinal Cord , Aging/genetics , Aging/metabolism , Aging/pathology , Autopsy , Cell Survival , Female , Humans , Male , Motor Neurons/metabolism , Motor Neurons/pathology , Muscular Atrophy, Spinal/drug therapy , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , Spinal Cord/metabolism , Spinal Cord/pathology , Survival of Motor Neuron 2 Protein/antagonists & inhibitors , Survival of Motor Neuron 2 Protein/genetics , Survival of Motor Neuron 2 Protein/metabolism
7.
Appl Microbiol Biotechnol ; 103(16): 6543-6557, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31236615

ABSTRACT

Lymphocyte activation gene-3 (LAG3) is a transmembrane protein expressed on activated T cells and delivers inhibitory signals to render the T cells unable to effectively help B cells to produce antibodies to microbes and vaccines. Presumably, antagonizing LAG3 could enhance the antibody responses to vaccines, and LAG3 antagonists could facilitate vaccines to induce vigorous antibody responses. In this study, we designed a LAG3-interfering antisense oligonucleotide, designated as LIO-1. The LIO-1 is complementary to an identical region shared in human and mouse LAG3 mRNA. We demonstrated that LIO-1 induced the degradation of LAG3 mRNA in immune cells, decreased the LAG3 expression on CD4+ T cells, maintained the prolonged proliferation and promoted the activation of antigen-specific CD4+ T cells, and increased the production of IFN-γ, IL-2, and IL-6 in the antigen re-stimulated immune cells. In addition, we found that LIO-1 enhanced the antibody responses induced by ISA35-formulated recombinant antigen vaccine or ISA35-formulated inactivated influenza virus vaccines in mice. Thus, the LIO-1, a nucleic acid LAG3 antagonist, could facilitate vaccines to induce vigorous antibody responses and has the possibility to be used as a novel adjuvant.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antibodies, Viral/blood , Antibody Formation , Antigens, CD/biosynthesis , Influenza Vaccines/immunology , Oligodeoxyribonucleotides, Antisense/administration & dosage , Recombinant Proteins/immunology , Animals , Down-Regulation , Influenza Vaccines/administration & dosage , Mice , Recombinant Proteins/administration & dosage , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Lymphocyte Activation Gene 3 Protein
8.
Adv Healthc Mater ; 8(5): e1801389, 2019 03.
Article in English | MEDLINE | ID: mdl-30714345

ABSTRACT

Tumor necrosis factor alpha (TNF-α) is usually regarded as a potential target for inflammatory bowel disease therapy. Herein, a promising strategy for effective delivery of phosphorothioated antisense oligodeoxyribonucleotide of TNF-α (PS-ATNF-α), targeting the intestinal inflammation based on the interaction of the single chain of triple helical ß-glucan (s-LNT) with poly-deoxyadenylic acid [poly(dA)], and the colon-specific degradation of chitosan-alginate (CA) hydrogel, is reported. The target gene of PS-ATNF-α, with a poly(dA) tail through a disulfide bond (-SS-), interacts with s-LNT to form a rod-like nanocomposite of s-LNT/poly(dA)-SS-PS-ATNF-α, which significantly inhibits lipopolysaccharide (LPS)-induced TNF-α at the protein level by 38.2% and mRNA level by 48.9% in RAW264.7 macrophages. The nanocomposites carried by the CA hydrogel with the loading amount of 83.5% are then orally administered and specifically released to the inflamed intestine, followed by internalization into intestinal cells such as macrophages, to reduce TNF-α production by 36.4% and dextran sulfate sodium-induced inflammation by decreasing myeloperoxidase and malondialdehyde. This study defines a new strategy for the oral delivery of antisense oligonucleotides to attenuate inflammatory response, demonstrating a notable potential for clinical applications in intestine-inflammation-targeted therapy.


Subject(s)
Inflammation/drug therapy , Intestines/drug effects , Nanocomposites/administration & dosage , Oligodeoxyribonucleotides, Antisense/administration & dosage , Polysaccharides/administration & dosage , Tumor Necrosis Factor-alpha/administration & dosage , Administration, Oral , Alginates/administration & dosage , Alginates/chemistry , Animals , Cell Line , Chitosan/administration & dosage , Chitosan/chemistry , Dextran Sulfate/pharmacology , Hydrogels/administration & dosage , Hydrogels/chemistry , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Nanocomposites/chemistry , Oligodeoxyribonucleotides, Antisense/chemistry , Polysaccharides/chemistry , RAW 264.7 Cells , Tumor Necrosis Factor-alpha/chemistry
9.
Mol Pharm ; 15(12): 5514-5520, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30350640

ABSTRACT

Mouse laser-induced choroidal neovascularization (mouse LCNV) recapitulates the "wet" form of human age-related macular degeneration (AMD). Vascular cell adhesion molecule-1 (VCAM-1) is a known inflammatory biomarker, and it increases in the choroidal neovascular tissues characteristic of this experimental model. We have designed and constructed gold nanoparticles (AuNPs) functionalized with hairpin-DNA that incorporates an antisense sequence complementary to VCAM-1 mRNA (AS-VCAM-1 hAuNPs) and tested them as optical imaging probes. The 3' end of the hairpin is coupled to a near-infrared fluorophore that is quenched by the AuNP surface via Förster resonance energy transfer (FRET). Hybridization of the antisense sequence to VCAM-1 mRNA displaces the fluorophore away from the AuNP surface, inducing fluorescent activity. In vitro testing showed that hAuNPs hybridize to an exogenous complementary oligonucleotide within a pH range of 4.5-7.4, and that they are stable at reduced pH. LCNV mice received tail-vein injections of AS-VCAM-1 hAuNPs. Hyperspectral imaging revealed the delivery of AS-VCAM-1 hAuNPs to excised choroidal tissues. Fluorescent images of CNV lesions were obtained, presumably in response to the hybridization of AS-hAuNPs to LCNV-induced VCAM-1 mRNA. This is the first demonstration of systemic delivery of hAuNPs to ocular tissues to facilitate mRNA imaging of any target.


Subject(s)
Choroidal Neovascularization/diagnostic imaging , Molecular Probes/administration & dosage , RNA, Messenger/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Wet Macular Degeneration/diagnostic imaging , Animals , Biomarkers/metabolism , Choroid/blood supply , Choroid/diagnostic imaging , Choroid/pathology , Choroid/radiation effects , Choroidal Neovascularization/etiology , Choroidal Neovascularization/pathology , Disease Models, Animal , Fluorescent Dyes/administration & dosage , Fluorescent Dyes/chemistry , Gold/administration & dosage , Gold/chemistry , Humans , Intravital Microscopy/methods , Lasers/adverse effects , Male , Metal Nanoparticles/administration & dosage , Metal Nanoparticles/chemistry , Mice , Mice, Inbred C57BL , Molecular Imaging/methods , Molecular Probes/chemistry , Oligodeoxyribonucleotides, Antisense/administration & dosage , Oligodeoxyribonucleotides, Antisense/chemistry , Optical Imaging/methods , Vascular Cell Adhesion Molecule-1/genetics , Wet Macular Degeneration/etiology , Wet Macular Degeneration/pathology
10.
Nucleic Acids Res ; 46(20): 10905-10916, 2018 11 16.
Article in English | MEDLINE | ID: mdl-30169779

ABSTRACT

Progress in oligonucleotide chemistry has produced a shift in the nature of siRNA used, from formulated, minimally modified siRNAs, to unformulated, heavily modified siRNA conjugates. The introduction of extensive chemical modifications is essential for conjugate-mediated delivery. Modifications have a significant impact on siRNA efficacy through interference with recognition and processing by RNAi enzymatic machinery, severely restricting the sequence space available for siRNA design. Many algorithms available publicly can successfully predict the activity of non-modified siRNAs, but the efficiency of the algorithms for designing heavily modified siRNAs has never been systematically evaluated experimentally. Here we screened 356 cholesterol-conjugated siRNAs with extensive modifications and developed a linear regression-based algorithm that effectively predicts siRNA activity using two independent datasets. We further demonstrate that predictive determinants for modified and non-modified siRNAs differ substantially. The algorithm developed from the non-modified siRNAs dataset has no predictive power for modified siRNAs and vice versa. In the context of heavily modified siRNAs, the introduction of chemical asymmetry fully eliminates the requirement for thermodynamic bias, the major determinant for non-modified siRNA efficacy. Finally, we demonstrate that in addition to the sequence of the target site, the accessibility of the neighboring 3' region significantly contributes to siRNA efficacy.


Subject(s)
Cholesterol/chemistry , RNA Interference , RNA Processing, Post-Transcriptional , RNA, Small Interfering/genetics , Base Sequence , Gene Expression Regulation , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Oligodeoxyribonucleotides, Antisense/administration & dosage , Oligodeoxyribonucleotides, Antisense/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/chemistry , Sequence Homology, Nucleic Acid , Thermodynamics
11.
Methods Mol Biol ; 1828: 381-394, 2018.
Article in English | MEDLINE | ID: mdl-30171555

ABSTRACT

Antisense oligonucleotides (AONs) have been actively developed for more than 30 years as a form of molecular medicine and represent promising therapeutic tools for many disorders. Significant progress has been made toward their clinical development in particular for splice switching AONs for the treatment of neuromuscular disorders such as Duchenne muscular dystrophy (DMD). Many different chemistries of AONs can be used for splice switching modulation, and some of them have now reached regulatory approval. However, despite advances in AON chemistry and design, systemic use of AONs is limited due to poor tissue uptake and sufficient therapeutic efficacy is difficult to achieve. Therefore, there is still a critical need to develop efficient AONs able to target all relevant tissues and international efforts are currently on going to advance new compounds or alternative chemistries with higher therapeutic potential. Here we describe the methods to evaluate the potency of tricyclo-DNA (tcDNA)-AONs, a novel class of AONs which displays unique pharmacological properties and unprecedented uptake in many tissues after systemic administration (Goyenvalle et al., Nat Med 21:270-275, 2015; Goyenvalle et al., J Neuromuscul Dis 3:157-167, 2016; Relizani et al., Mol Ther Nucleic Acids 8:144-157, 2017; Robin et al., Mol Ther Nucleic Acids 7:81-89, 2017). We will focus on the preclinical evaluation of these tcDNA for DMD, specifically targeting the exon 51 of the human dystrophin gene. We will first detail methods to analyze their efficacy both in vitro in human myoblasts and in vivo in the hDMD and mdx52 mouse models and then describe means to evaluate their potential renal toxicity.


Subject(s)
Dystrophin/genetics , Exons , Muscular Dystrophy, Duchenne/genetics , Oligodeoxyribonucleotides, Antisense/chemistry , Oligodeoxyribonucleotides, Antisense/genetics , RNA Splicing , Animals , Biomarkers , Cells, Cultured , Gene Targeting , Humans , Kidney/metabolism , Liver/metabolism , Mice , Myoblasts/metabolism , Oligodeoxyribonucleotides, Antisense/administration & dosage , Transfection
12.
Theranostics ; 8(7): 1923-1939, 2018.
Article in English | MEDLINE | ID: mdl-29556365

ABSTRACT

Conventional chemotherapy of pancreatic cancer (PaCa) suffers the problems of low drug permeability and inherent or acquired drug resistance. Development of new strategies for enhanced therapy still remains a great challenge. Herein, we report a new ultrasound-targeted microbubble destruction (UTMD)-promoted delivery system based on dendrimer-entrapped gold nanoparticles (Au DENPs) for co-delivery of gemcitabine (Gem) and miR-21 inhibitor (miR-21i). Methods: In this study, Gem-Au DENPs/miR-21i was designed and synthesized. The designed polyplexes were characterized via transmission electron microscopy (TEM), Gel retardation assay and dynamic light scattering (DLS). Then, the optimum exposure parameters were examined by an ultrasound exposure platform. The cellular uptake, cytotoxicity and anticancer effects in vitro were analyzed by confocal laser microscopy, spectra microplate reader, flow cytometry and a chemiluminescence imaging system. Lastly, the anticancer effects in vivo were evaluated by contrast-enhanced ultrasound (CEUS), hematoxylin and eosin (H&E) staining, TUNEL staining and comparison of tumor volume. Results: The results showed that the Gem-Au DENPs/miR-21i can be uptake by cancer cells and the cellular uptake was further facilitated by UTMD with an ultrasound power of 0.4 W/cm2 to enhance the cell permeability. Further, the co-delivery of Gem and miR-21i with or without UTMD treatment displayed 82-fold and 13-fold lower IC50 values than the free Gem, respectively. The UTMD-promoted co-delivery of Gem and miR-21i was further validated by in vivo treatment and showed a significant tumor volume reduction and an increase in blood perfusion of xenografted pancreatic tumors. Conclusion: The co-delivery of Gem and miR-21i using Au DENPs can be significantly promoted by UTMD technology, hence providing a promising strategy for effective pancreatic cancer treatments.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Deoxycytidine/analogs & derivatives , Metal Nanoparticles/administration & dosage , Molecular Targeted Therapy/methods , Oligodeoxyribonucleotides, Antisense/administration & dosage , Pancreatic Neoplasms/drug therapy , Ultrasonography , Animals , Cell Line, Tumor , Cell Survival/drug effects , Dendrimers/administration & dosage , Deoxycytidine/administration & dosage , Drug Carriers/administration & dosage , Gold/administration & dosage , Heterografts , Humans , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/antagonists & inhibitors , Microbubbles , Neoplasm Transplantation , Pancreatic Neoplasms/diagnostic imaging , Treatment Outcome , Gemcitabine
13.
Neuropharmacology ; 133: 254-263, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29407215

ABSTRACT

Hydrogen sulfide (H2S) formed by cystathionine-γ-lyase (CSE) enhances the activity of Cav3.2 T-type Ca2+ channels, contributing to the bladder pain accompanying hemorrhagic cystitis caused by systemic administration of cyclophosphamide (CPA) in mice. Given clinical and fundamental evidence for the involvement of the substance P/NK1 receptor systems in bladder pain syndrome (BPS)/interstitial cystitis (IC), we created an intravesical substance P-induced bladder pain model in mice and analyzed the possible involvement of the CSE/Cav3.2 pathway. Bladder pain/cystitis was induced by i.p. CPA or intravesical substance P in female mice. Bladder pain was evaluated by counting nociceptive behavior and by detecting referred hyperalgesia in the lower abdomen and hindpaw. The isolated bladder tissue was weighed to estimate bladder swelling and subjected to histological observation and Western blotting. Intravesical substance P caused profound referred hyperalgesia accompanied by little bladder swelling or edema 6-24 h after the administration, in contrast to i.p. CPA-induced nociceptive behavior/referred hyperalgesia with remarkable bladder swelling/edema and urothelial damage. The bladder pain and/or cystitis symptoms caused by substance P or CPA were prevented by the NK1 receptor antagonist. CSE in the bladder was upregulated by substance P or CPA, and the NK1 antagonist prevented the CPA-induced CSE upregulation. A CSE inhibitor, a T-type Ca2+ channel blocker and gene silencing of Cav3.2 abolished the intravesical substance P-induced referred hyperalgesia. The intravesical substance P-induced pain in mice is useful as a model for nonulcerative BPS, and involves the activation of the NK1 receptor/CSE/H2S/Cav3.2 cascade.


Subject(s)
Calcium Channels, T-Type/metabolism , Cystathionine gamma-Lyase/metabolism , Pain/etiology , Signal Transduction/drug effects , Substance P/toxicity , Urinary Bladder Diseases/chemically induced , Urinary Bladder Diseases/complications , Animals , Benzimidazoles/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/genetics , Cyclophosphamide/toxicity , Cyclopropanes/pharmacology , Cystathionine gamma-Lyase/genetics , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Immunosuppressive Agents/toxicity , Mice , Naphthalenes/pharmacology , Oligodeoxyribonucleotides, Antisense/administration & dosage , Pain/chemically induced , Pain Measurement , RNA, Messenger/metabolism , Receptors, Neurokinin-1/genetics , Receptors, Neurokinin-1/metabolism , Spinal Cord/pathology , Urinary Bladder Diseases/pathology , Urothelium/drug effects , Urothelium/pathology
14.
Diabetes Care ; 41(4): 807-814, 2018 04.
Article in English | MEDLINE | ID: mdl-29439147

ABSTRACT

OBJECTIVE: To evaluate safety and efficacy of IONIS-PTP-1BRx, a second-generation 2'-O-methoxyethyl antisense inhibitor of protein tyrosine phosphatase 1B, as add-on therapy in overweight patients with type 2 diabetes inadequately controlled with metformin with or without sulfonylurea therapy. RESEARCH DESIGN AND METHODS: In this phase II, double-blind, randomized, placebo-controlled, multicenter trial, overweight and obese patients (BMI ≥27 kg/m2) with type 2 diabetes (HbA1c ≥7.5% [58 mmol/mol] and ≤10.5% [91 mmol/mol]) on a stable dose of metformin alone or with sulfonylurea were randomized 2:1 to IONIS-PTP-1BRx 200 mg (n = 62) or placebo (n = 30) once weekly for 26 weeks. RESULTS: Mean baseline HbA1c was 8.6% (70 mmol/mol) and 8.7% (72 mmol/mol) in placebo and active treatment, respectively. At week 27, IONIS-PTP-1BRx reduced mean HbA1c levels by -0.44% (-4.8 mmol/mol; P = 0.074) from baseline and improved leptin (-4.4 ng/mL; P = 0.007) and adiponectin (0.99 µg/mL; P = 0.026) levels compared with placebo. By week 36, mean HbA1c was significantly reduced (-0.69% [-7.5 mmol/mol]; P = 0.034) and accompanied by reductions in fructosamine (-33.2 µmol/L; P = 0.005) and glycated albumin (-1.6%; P = 0.031) versus placebo. Despite both treatment groups receiving similar lifestyle counseling, mean body weight significantly decreased from baseline to week 27 with IONIS-PTP-1BRx versus placebo (-2.6 kg; P = 0.002) independent of HbA1c reduction (R2 = 0.0020). No safety concerns were identified in the study. CONCLUSIONS: Compared with placebo, IONIS-PTP-1BRx treatment for 26 weeks produced prolonged reductions in HbA1c, improved medium-term glycemic parameters, reduced leptin and increased adiponectin levels, and resulted in a distinct body weight-reducing effect.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Insulin Resistance , Obesity/drug therapy , Oligodeoxyribonucleotides, Antisense/therapeutic use , Oligodeoxyribonucleotides/therapeutic use , Overweight/drug therapy , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Weight Loss/drug effects , Adult , Aged , Blood Glucose/drug effects , Blood Glucose/metabolism , Body Weight/drug effects , Body Weight/genetics , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Double-Blind Method , Drug Therapy, Combination , Female , Humans , Hypoglycemic Agents/administration & dosage , Insulin Resistance/genetics , Male , Metformin/administration & dosage , Middle Aged , Obesity/complications , Obesity/metabolism , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides, Antisense/administration & dosage , Overweight/complications , Overweight/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Sulfonylurea Compounds/administration & dosage , Weight Loss/genetics
15.
Brain Behav Immun ; 72: 34-44, 2018 08.
Article in English | MEDLINE | ID: mdl-29128611

ABSTRACT

p38 mitogen-activated protein kinase (MAPK) consists of two major isoforms: p38α and p38ß; however, it remains unclear which isoform is more important for chronic pain development. Recently, we developed potent, long-lasting, and p38 MAPK subtype-specific antisense oligonucleotides (ASOs). We examined the therapeutic effects of isoform-specific ASOs in several chronic pain models following single intrathecal injection (300 µg/10 µl) in CD1 mice. In the chronic constriction injury (CCI) model, p38α MAPK ASO, given on post-operative day 5, reduced CCI-induced mechanical allodynia in male but not female mice. In contrast, mechanical allodynia after CCI in both sexes was not affected by p38ß MAPK ASO. Intrathecal injection of p38α or p38ß ASO resulted in a partial reduction (≈ 50%) of spinal p38α or p38ß mRNA level, respectively, in both sexes at two weeks. In contrast, intrathecal injection of the ASOs did not affect p38α and p38ß MAPK mRNA levels in dorsal root ganglia. Intrathecal p38α ASO also reduced postoperative pain (mechanical and cold allodynia) in male mice after tibia fracture. However, intrathecal p38α ASO had no effect on mechanical allodynia in male mice after paclitaxel treatment. Intrathecal p38α MAPK ASO pre-treatment also prevented TLR4-mediated mechanical allodynia and downregulated levels of p38α MAPK and phosphorylated p38 MAPK following intrathecal treatment of lipopolysaccharide. In summary, our findings suggest that p38α MAPK is the major p38 MAPK isoform in the spinal cord and regulates chronic pain in a sex and model-dependent manner. Intrathecal p38α MAPK ASO may offer a new treatment for some chronic pain conditions.


Subject(s)
Neuralgia/therapy , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Disease Models, Animal , Down-Regulation , Female , Ganglia, Spinal/metabolism , Hyperalgesia/chemically induced , Hyperalgesia/therapy , Injections, Spinal , Male , Mice , Microglia/metabolism , Mitogen-Activated Protein Kinase 11/genetics , Mitogen-Activated Protein Kinase 11/metabolism , Mitogen-Activated Protein Kinase 14/genetics , Mitogen-Activated Protein Kinase 14/metabolism , Oligodeoxyribonucleotides, Antisense/administration & dosage , Oligodeoxyribonucleotides, Antisense/genetics , Pain Measurement , Pain, Postoperative/therapy , Peripheral Nervous System Diseases/metabolism , Phosphorylation , Protein Isoforms , Spinal Cord/metabolism , Toll-Like Receptor 4/metabolism
16.
Mol Cells ; 40(3): 186-194, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28301916

ABSTRACT

A brain-enriched secreting signal peptide, NELL2, has been suggested to play multiple roles in the development, survival, and activity of neurons in mammal. We investigated here a possible involvement of central NELL2 in regulating feeding behavior and metabolism. In situ hybridization and an im-munohistochemical approach were used to determine expression of NELL2 as well as its colocalization with proopiomelanocortin (POMC) and neuropeptide Y (NPY) in the rat hypothalamus. To investigate the effect of NELL2 on feeding behavior, 2 nmole of antisense NELL2 oligodeoxynucleotide was administered into the lateral ventricle of adult male rat brains for 6 consecutive days, and changes in daily body weight, food, and water intake were monitored. Metabolic state-dependent NELL2 expression in the hypothalamus was tested in vivo using a fasting model. NELL2 was noticeably expressed in the hypothalamic nuclei controlling feeding behavior. Furthermore, all arcuatic POMC and NPY positive neurons produced NELL2. The NELL2 gene expression in the hypothalamus was up-regulated by fasting. However, NELL2 did not affect POMC and NPY gene expression in the hypothalamus. A blockade of NELL2 production in the hypothalamus led to a reduction in daily food intake, followed by a loss in body weight without a change in daily water intake in normal diet condition. NELL2 did not affect short-term hunger dependent appetite behavior. Our data suggests that hypothalamic NELL2 is associated with appetite behavior, and thus central NELL2 could be a new therapeutic target for obesity.


Subject(s)
Feeding Behavior/drug effects , Hypothalamus/metabolism , Nerve Tissue Proteins/metabolism , Oligodeoxyribonucleotides, Antisense/administration & dosage , Animals , Body Weight/drug effects , Eating/drug effects , Fasting/metabolism , Male , Nerve Tissue Proteins/antagonists & inhibitors , Neuropeptide Y/metabolism , Pro-Opiomelanocortin/metabolism , Rats , Up-Regulation
17.
Bioorg Med Chem ; 24(1): 26-32, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26678173

ABSTRACT

The targeting of abundant hepatic asialoglycoprotein receptors (ASGPR) with trivalent N-acetylgalactosamine (GalNAc) is a reliable strategy for efficiently delivering antisense oligonucleotides (ASOs) to the liver. We here experimentally demonstrate the high systemic potential of the synthetically-accessible, phosphodiester-linked monovalent GalNAc unit when tethered to the 5'-terminus of well-characterised 2',4'-bridged nucleic acid (also known as locked nucleic acid)-modified apolipoprotein B-targeting ASO via a bio-labile linker. Quantitative analysis of the hepatic disposition of the ASOs revealed that phosphodiester is preferable to phosphorothioate as an interunit linkage in terms of ASGPR binding of the GalNAc moiety, as well as the subcellular behavior of the ASO. The flexibility of this monomeric unit was demonstrated by attaching up to 5 GalNAc units in a serial manner and showing that knockdown activity improves as the number of GalNAc units increases. Our study suggests the structural requirements for efficient hepatocellular targeting using monovalent GalNAc and could contribute to a new molecular design for suitably modifying ASO.


Subject(s)
Acetylgalactosamine/analogs & derivatives , Acetylgalactosamine/chemical synthesis , Hepatocytes/metabolism , Liver/metabolism , Oligodeoxyribonucleotides, Antisense/chemical synthesis , Organophosphorus Compounds/chemical synthesis , Alanine Transaminase/blood , Animals , Apolipoproteins B/genetics , Apolipoproteins B/metabolism , Asialoglycoprotein Receptor/metabolism , Aspartate Aminotransferases/blood , Cholesterol/blood , Gene Knockdown Techniques , Gene Silencing , Hepatocytes/drug effects , Hepatocytes/pathology , Liver/drug effects , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Oligodeoxyribonucleotides, Antisense/administration & dosage , Oligodeoxyribonucleotides, Antisense/toxicity , RNA, Messenger/metabolism
18.
Med Sci Monit ; 21: 2339-44, 2015 Aug 10.
Article in English | MEDLINE | ID: mdl-26258995

ABSTRACT

BACKGROUND: To investigate the in vitro inhibitory effects of PEI-RGD/125I-(αv)ASODN (PEI, polyethylenimine; RGD, Arg-Gly-Asp; ASODN, antisense oligodeoxynucleotide) on the growth and invasion of HepG2 cells. MATERIAL AND METHODS: ASODN of the integrin αv-subunit was marked with 125I and underwent complexation with PEI-RGD, a PEI derivative. Next, PEI-RGD/125I-(αv) ASODN was introduced into HepG2 cells via receptor-mediated transfection, and its inhibition rate on HepG2 cell growth was tested using the methyl thiazolyl tetrazolium (MTT) method. The effects of PEI-RGD/125I-(αv) ASODN on HepG2 cell invasion ability were evaluated using the Boyden chamber assay. RESULTS: 1) The 125I marking rate of (αv) ASODN was 73.78±4.09%, and the radiochemical purity was 96.68±1.38% (greater than 90% even after a 48-h incubation period at 37°C), indicating high stability. 2) The cytotoxicity assays showed that the cell inhibition rates did not differ significantly between the PEI-RGD/125I-(αv)ASODN group and the PEI-RGD/(αv) ASODN group, but they were both significantly higher than in the other groups and were positively correlated (r=0.879) with the dosage within a certain range. 3) The invasion assays showed that the inhibition rate was significantly greater in the PEI-RGD/125I-(αv) ASODN group compared to the other groups. CONCLUSIONS: PEI-RGD/125I-(αv) ASODN can efficiently inhibit the growth and proliferation of HepG2 cells and can also weaken their invasive ability.


Subject(s)
Integrin alphaV/genetics , Iodine Radioisotopes/administration & dosage , Oligodeoxyribonucleotides, Antisense/administration & dosage , Oligodeoxyribonucleotides, Antisense/genetics , Oligopeptides/administration & dosage , Polyethyleneimine/analogs & derivatives , Base Sequence , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Proliferation/radiation effects , Hep G2 Cells , Humans , Neoplasm Invasiveness , Polyethyleneimine/administration & dosage
19.
Neuropharmacology ; 93: 116-23, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25656477

ABSTRACT

Previously we demonstrated that exposure of the central nucleus of the amygdala (CeA) to elevated corticosterone (CORT) induces nociceptive behaviors that are reversed by glucocorticoid and/or mineralocorticoid (GR/MR) receptor antagonism. Here we test the hypothesis that in a cholesterol (CHOL)-implanted control rat, selective knockdown of GR/MR in the CeA would, via a corticotropin-releasing factor (CRF)-mediated mechanism, replicate the nociceptive behaviors produced by elevated amygdala CORT. Micropellets of CHOL or CORT were stereotaxically placed on the dorsal margin of the CeA. Cannulae were implanted into the CeA for the delivery of vehicle or oligodeoxynucleotide (ODN) of either antisense (ASO) or random sequences (RSO) targeting GR or MR. Visceromotor behavioral response quantified visceral sensitivity in response to colonic distension, while von Frey filaments assessed somatic sensitivity. Receptor expression was determined with qRT-PCR. In CHOL implanted controls, knockdown of GR in the CeA increased both colonic and somatic sensitivity, whereas selective knockdown of MR in the CeA induced colonic hypersensitivity without affecting somatic sensitivity. CRF expression in the CeA was increased in CHOL-implanted rats treated with GR or MR ASO and resembled the augmented CRF expression seen in the CORT-implanted rats. This is the first study to demonstrate that decreasing either GR or MR within the CeA is sufficient to induce visceral hypersensitivity whereas somatic hypersensitivity developed after only GR knockdown. The loss of either GR or MR was associated with an increased CRF expression, and may represent a common mechanism for the development of CeA-mediated nociceptive behaviors.


Subject(s)
Central Amygdaloid Nucleus/physiology , Corticosterone/metabolism , Nociception/physiology , Pain/etiology , Receptors, Glucocorticoid/deficiency , Analysis of Variance , Animals , Central Amygdaloid Nucleus/drug effects , Cholesterol/administration & dosage , Corticosterone/genetics , Male , Mineralocorticoids/metabolism , Nociception/drug effects , Oligodeoxyribonucleotides, Antisense/administration & dosage , Physical Stimulation/adverse effects , Rats , Rats, Inbred F344 , Receptors, Glucocorticoid/genetics , Receptors, Mineralocorticoid/deficiency , Receptors, Mineralocorticoid/genetics , Visceral Pain/etiology
20.
Cancer Immunol Immunother ; 64(4): 447-57, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25579379

ABSTRACT

Autologous glioblastoma multiforme tumor cells treated with an antisense oligodeoxynucleotide (AS-ODN) targeting insulin-like growth factor receptor-1 (IGF-1R) are the basis of a vaccine with therapeutic effects on tumor recurrence in a pilot clinical trial. As a preface to continued clinical investigation of this vaccination strategy, we have studied the contribution of an optimized IGF-1R AS-ODN, designated "NOBEL", to the induction of immunity to mouse GL261 glioma cells. The impact of NOBEL on mechanisms contributing to the development of GL261 immunity was first examined in the periphery. GL261 cells are naturally immunogenic when implanted into the flanks of congenic C57BL/6 mice, immunizing rather than forming tumors in around 50 % of these animals but causing tumors in the majority of mice lacking T and B lymphocytes. Overnight treatment with NOBEL in vitro reduces IGF-1R expression by GL261 cells but has minimal effect on cell viability and does not reduce the capacity of the cells to form tumors upon implantation. In contrast, tumors are extremely rare when GL261 cells are mixed with NOBEL at inoculation into the flanks of C57BL/6, and the recipient mice become immune to subcutaneous and intracranial challenge with untreated GL261. Adaptive immune mechanisms contribute to this effect, as immunocompromised mice fail to either fully control tumor formation or develop immunity following flank administration of the GL261/NOBEL mix. NOBEL's structure has known immunostimulatory motifs that likely contribute to the immunogenicity of the mix, but its specificity for IGF-1R mRNA is also important as a similarly structured sense molecule is not effective.


Subject(s)
Brain Neoplasms/immunology , Glioma/immunology , Immunity, Cellular/immunology , Immunotherapy , Oligodeoxyribonucleotides, Antisense/administration & dosage , Receptor, IGF Type 1/immunology , Animals , Blotting, Western , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Glioma/pathology , Glioma/therapy , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligodeoxyribonucleotides, Antisense/genetics , Oligodeoxyribonucleotides, Antisense/immunology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor, IGF Type 1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , beta 2-Microglobulin/physiology
SELECTION OF CITATIONS
SEARCH DETAIL