Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters

Publication year range
1.
J Am Chem Soc ; 143(9): 3340-3347, 2021 03 10.
Article in English | MEDLINE | ID: mdl-33648338

ABSTRACT

Floxuridine oligomers are anticancer oligonucleotide drugs composed of a number of floxuridine residues. They show enhanced cytotoxicity per floxuridine monomer because the nuclease degradation of floxuridine oligomers directly releases highly active floxuridine monophosphate in cells. However, their clinical use is limited by the low selectivity against cancer cells. To address this limitation, we herein report floxuridine oligomer prodrugs that are active under hypoxia conditions, which is one of the distinguishing features of the microenvironment of all solid tumors. We designed and synthesized two types of floxuridine oligomer prodrugs that possess hypoxia-responsive moieties on nucleobases. The floxuridine oligomer prodrugs showed lower cytotoxicity under normoxia conditions (O2 = 20%), while the parent floxuridine oligomer showed similar anticancer effects under hypoxia conditions (O2 = 1%). The floxuridine oligomer prodrug enabled tumor growth suppression in live mice. This would be the first example demonstrating the conditional control of the medicinal efficacy of oligomerized nucleoside anticancer drugs.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Floxuridine/analogs & derivatives , Floxuridine/therapeutic use , Neoplasms/drug therapy , Oligoribonucleotides/therapeutic use , Prodrugs/therapeutic use , Animals , Cell Line, Tumor , Humans , Hypoxia/physiopathology , Mice, Inbred BALB C , Mice, Nude , Neoplasms/physiopathology , Xenograft Model Antitumor Assays
2.
Int J Mol Sci ; 19(2)2018 Feb 03.
Article in English | MEDLINE | ID: mdl-29401684

ABSTRACT

Acute myeloid leukemias (AML) are clonal disorders of hematopoietic progenitor cells which are characterized by relevant heterogeneity in terms of phenotypic, genotypic, and clinical features. Among the genetic aberrations that control disease development there are microRNAs (miRNAs). miRNAs are small non-coding RNAs that regulate, at post-transcriptional level, translation and stability of mRNAs. It is now established that deregulated miRNA expression is a prominent feature in AML. Functional studies have shown that miRNAs play an important role in AML pathogenesis and miRNA expression signatures are associated with chemotherapy response and clinical outcome. In this review we summarized miRNA signature in AML with different cytogenetic, molecular and clinical characteristics. Moreover, we reviewed the miRNA regulatory network in AML pathogenesis and we discussed the potential use of cellular and circulating miRNAs as biomarkers for diagnosis and prognosis and as therapeutic targets.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/therapy , MicroRNAs/genetics , Oncogene Proteins, Fusion/genetics , Animals , Antagomirs/genetics , Antagomirs/metabolism , Antagomirs/therapeutic use , Biomarkers, Tumor/agonists , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/metabolism , Chromosome Aberrations , Extracellular Vesicles/metabolism , Extracellular Vesicles/pathology , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice , MicroRNAs/agonists , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Molecular Targeted Therapy , Oligoribonucleotides/genetics , Oligoribonucleotides/metabolism , Oligoribonucleotides/therapeutic use , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/metabolism , Prognosis , Signal Transduction , Xenograft Model Antitumor Assays
3.
Int J Cancer ; 139(7): 1648-57, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27194579

ABSTRACT

The eukaryotic translation initiation factor 4E (eIF4E) is a potent oncogene that is found to be dysregulated in 30% of human cancer, including colorectal carcinogenesis (CRC). ISIS 183750 is a second-generation antisense oligonucleotide (ASO) designed to inhibit the production of the eIF4E protein. In preclinical studies we found that EIF4e ASOs reduced expression of EIF4e mRNA and inhibited proliferation of colorectal carcinoma cells. An additive antiproliferative effect was observed in combination with irinotecan. We then performed a clinical trial evaluating this combination in patients with refractory cancer. No dose-limiting toxicities were seen but based on pharmacokinetic data and tolerability the dose of irinotecan was reduced to 160 mg/m(2) biweekly. Efficacy was evaluated in 15 patients with irinotecan-refractory colorectal cancer. The median time of disease control was 22.1 weeks. After ISIS 183750 treatment, peripheral blood levels of eIF4E mRNA were decreased in 13 of 19 patients. Matched pre- and posttreatment tumor biopsies showed decreased eIF4E mRNA levels in five of nine patients. In tumor tissue, the intracellular and stromal presence of ISIS 183750 was detected by IHC in all biopsied patients. Although there were no objective responses stable disease was seen in seven of 15 (47%) patients who were progressing before study entry, six of whom were stable at the time of the week 16 CT scan. We were also able to confirm through mandatory pre- and posttherapy tumor biopsies penetration of the ASO into the site of metastasis.


Subject(s)
Camptothecin/analogs & derivatives , Colorectal Neoplasms/therapy , Eukaryotic Initiation Factor-4E/antagonists & inhibitors , Oligonucleotides, Antisense/therapeutic use , Oligoribonucleotides/therapeutic use , Adult , Aged , Camptothecin/adverse effects , Camptothecin/blood , Camptothecin/therapeutic use , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Combined Modality Therapy , Eukaryotic Initiation Factor-4E/genetics , Female , HCT116 Cells , Humans , Irinotecan , Male , Middle Aged , Oligonucleotides , Oligonucleotides, Antisense/genetics , Oligoribonucleotides/genetics , RNA, Messenger/blood , RNA, Messenger/genetics
4.
Blood ; 124(17): 2643-6, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25163699

ABSTRACT

Increased hepcidin production is key to the development of anemia of inflammation. We investigated whether lexaptepid, an antihepcidin l-oligoribonucleotide, prevents the decrease in serum iron during experimental human endotoxemia. This randomized, double-blind, placebo-controlled trial was carried out in 24 healthy males. At T = 0 hours, 2 ng/kg Escherichia coli lipopolysaccharide was intravenously administered, followed by an intravenous injection of 1.2 mg/kg lexaptepid or placebo at T = 0.5 hours. The lipopolysaccharide-induced inflammatory response was similar in subjects treated with lexaptepid or placebo regarding clinical and biochemical parameters. At T = 9 hours, serum iron had increased by 15.9 ± 9.8 µmol/L from baseline in lexaptepid-treated subjects compared with a decrease of 8.3 ± 9.0 µmol/L in controls (P < .0001). This study delivers proof of concept that lexaptepid achieves clinically relevant hepcidin inhibition enabling investigations in the treatment of anemia of inflammation. This trial was registered at www.clinicaltrial.gov as #NCT01522794.


Subject(s)
Inflammation/blood , Inflammation/prevention & control , Iron/blood , Oligoribonucleotides/therapeutic use , Adolescent , Adult , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/therapeutic use , C-Reactive Protein/metabolism , Double-Blind Method , Endotoxemia/blood , Endotoxemia/chemically induced , Endotoxemia/prevention & control , Hepcidins/antagonists & inhibitors , Hepcidins/blood , Humans , Inflammation/chemically induced , Injections, Intravenous , Interleukin 1 Receptor Antagonist Protein/blood , Interleukin-10 , Interleukin-6/blood , Leukocyte Count , Lipopolysaccharides , Male , Metabolic Clearance Rate , Oligoribonucleotides/administration & dosage , Oligoribonucleotides/pharmacokinetics , Treatment Outcome , Tumor Necrosis Factor-alpha/blood , Young Adult
5.
Blood ; 121(12): 2311-5, 2013 Mar 21.
Article in English | MEDLINE | ID: mdl-23349391

ABSTRACT

Anemia of chronic inflammation is the most prevalent form of anemia in hospitalized patients. A hallmark of this disease is the intracellular sequestration of iron. This is a consequence of hepcidin-induced internalization and subsequent degradation of ferroportin, the hepcidin receptor and only known iron-export protein. This study describes the characterization of novel anti-hepcidin compound NOX-H94, a structured L-oligoribonucleotide that binds human hepcidin with high affinity (Kd = 0.65 ± 0.06 nmol/L). In J774A.1 macrophages, NOX-H94 blocked hepcidin-induced ferroportin degradation and ferritin expression (half maximal inhibitory concentration = 19.8 ± 4.6 nmol/L). In an acute cynomolgus monkey model of interleukin 6 (IL-6)-induced hypoferremia, NOX-H94 inhibited serum iron reduction completely. In a subchronic model of IL-6-induced anemia, NOX-H94 inhibited the decrease in hemoglobin concentration. We conclude that NOX-H94 protects ferroportin from hepcidin-induced degradation. Therefore, this pharmacologic approach may represent an interesting treatment option for patients suffering from anemia of chronic inflammation.


Subject(s)
Anemia/drug therapy , Anemia/etiology , Inflammation/complications , Inflammation/drug therapy , Oligoribonucleotides/therapeutic use , Anemia/pathology , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Antimicrobial Cationic Peptides/antagonists & inhibitors , Cells, Cultured , Disease Models, Animal , Drug Evaluation, Preclinical , Erythrocytes/drug effects , Erythrocytes/metabolism , Hemoglobins/analysis , Hemoglobins/drug effects , Hepcidins , Interleukin-6/administration & dosage , Interleukin-6/adverse effects , Iron/blood , Iron/metabolism , Iron Metabolism Disorders/chemically induced , Macaca fascicularis , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Oligoribonucleotides/administration & dosage , Oligoribonucleotides/pharmacology
6.
Am J Respir Crit Care Med ; 185(4): 409-19, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22161164

ABSTRACT

RATIONALE: MicroRNAs (miRs) control various cellular processes in tissue homeostasis and disease by regulating gene expression on the posttranscriptional level. Recently, it was demonstrated that the expression of miR-21 and members of the miR-17-92 cluster was significantly altered in experimental pulmonary hypertension (PH). OBJECTIVES: To evaluate the therapeutic efficacy and antiremodeling potential of miR inhibitors in the pathogenesis of PH. METHODS: We first tested the effects of miR inhibitors (antagomirs), which were specifically designed to block miR-17 (A-17), miR-21 (A-21), and miR-92a (A-92a) in chronic hypoxia-induced PH in mice and A-17 in monocrotaline-induced PH in rats. Moreover, biological function of miR-17 was analyzed in cultured pulmonary artery smooth muscle cells. MEASUREMENTS AND MAIN RESULTS: In the PH mouse model, A-17 and A-21 reduced right ventricular systolic pressure, and all antagomirs decreased pulmonary arterial muscularization. However, only A-17 reduced hypoxia-induced right ventricular hypertrophy and improved pulmonary artery acceleration time. In the monocrotaline-induced PH rat model, A-17 treatment significantly decreased right ventricular systolic pressure and total pulmonary vascular resistance index, increased pulmonary artery acceleration time, normalized cardiac output, and decreased pulmonary vascular remodeling. Among the tested miR-17 targets, the cyclin-dependent kinase inhibitor 1A (p21) was up-regulated in lungs undergoing A-17 treatment. Likewise, in human pulmonary artery smooth muscle cells, A-17 increased p21. Overexpression of miR-17 significantly reduced p21 expression and increased proliferation of smooth muscle cells. CONCLUSIONS: Our data demonstrate that A-17 improves heart and lung function in experimental PH by interfering with lung vascular and right ventricular remodeling. The beneficial effects may be related to the up-regulation of p21. Thus, inhibition of miR-17 may represent a novel therapeutic concept to ameliorate disease state in PH.


Subject(s)
Hypertension, Pulmonary/drug therapy , MicroRNAs/antagonists & inhibitors , MicroRNAs/physiology , Oligoribonucleotides/therapeutic use , Airway Remodeling/drug effects , Animals , Antagomirs , Blotting, Western , Cardiac Output/drug effects , Cells, Cultured , Disease Models, Animal , Humans , Hypertension, Pulmonary/metabolism , Mice , MicroRNAs/metabolism , Oligoribonucleotides/pharmacology , Pulmonary Artery/drug effects , Pulmonary Artery/physiology , Rats , Reverse Transcriptase Polymerase Chain Reaction , Vascular Resistance/drug effects , Ventricular Function, Right/drug effects
7.
ACS Biomater Sci Eng ; 9(3): 1656-1671, 2023 03 13.
Article in English | MEDLINE | ID: mdl-36853144

ABSTRACT

As the world braces to enter its fourth year of the coronavirus disease 2019 (COVID-19) pandemic, the need for accessible and effective antiviral therapeutics continues to be felt globally. The recent surge of Omicron variant cases has demonstrated that vaccination and prevention alone cannot quell the spread of highly transmissible variants. A safe and nontoxic therapeutic with an adaptable design to respond to the emergence of new variants is critical for transitioning to the treatment of COVID-19 as an endemic disease. Here, we present a novel compound, called SBCoV202, that specifically and tightly binds the translation initiation site of RNA-dependent RNA polymerase within the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) genome, inhibiting viral replication. SBCoV202 is a Nanoligomer, a molecule that includes peptide nucleic acid sequences capable of binding viral RNA with single-base-pair specificity to accurately target the viral genome. The compound has been shown to be safe and nontoxic in mice, with favorable biodistribution, and has shown efficacy against SARS-CoV-2 in vitro. Safety and biodistribution were assessed using three separate administration methods, namely, intranasal, intravenous, and intraperitoneal. Safety studies showed the Nanoligomer caused no outward distress, immunogenicity, or organ tissue damage, measured through observation of behavior and body weight, serum levels of cytokines, and histopathology of fixed tissue, respectively. SBCoV202 was evenly biodistributed throughout the body, with most tissues measuring Nanoligomer concentrations well above the compound KD of 3.37 nM. In addition to favorable availability to organs such as the lungs, lymph nodes, liver, and spleen, the compound circulated through the blood and was rapidly cleared through the renal and urinary systems. The favorable biodistribution and lack of immunogenicity and toxicity set Nanoligomers apart from other antisense therapies, while the adaptability of the nucleic acid sequence of Nanoligomers provides a defense against future emergence of drug resistance, making these molecules an attractive potential treatment for COVID-19.


Subject(s)
COVID-19 Drug Treatment , COVID-19 , Genome, Viral , Nanomedicine , Nanostructures , Oligoribonucleotides , Peptide Nucleic Acids , SARS-CoV-2 , SARS-CoV-2/drug effects , SARS-CoV-2/genetics , COVID-19/virology , COVID-19 Drug Treatment/adverse effects , COVID-19 Drug Treatment/methods , Nanostructures/administration & dosage , Nanostructures/adverse effects , Nanostructures/therapeutic use , Nanomedicine/methods , Patient Safety , Peptide Nucleic Acids/administration & dosage , Peptide Nucleic Acids/adverse effects , Peptide Nucleic Acids/pharmacokinetics , Peptide Nucleic Acids/therapeutic use , Oligoribonucleotides/administration & dosage , Oligoribonucleotides/adverse effects , Oligoribonucleotides/pharmacokinetics , Oligoribonucleotides/therapeutic use , Animals , Mice , Mice, Inbred BALB C , In Vitro Techniques , Genome, Viral/drug effects , Genome, Viral/genetics , Tissue Distribution
10.
Free Radic Biol Med ; 139: 1-15, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31102709

ABSTRACT

To protect tissues and the organism from disease, potentially harmful cells are removed through programmed cell death processes, including apoptosis and necroptosis. These types of cell death are critically controlled by microRNAs (miRNAs). MiRNAs are short RNA molecules that target and inhibit expression of many cellular regulators, including those controlling programmed cell death via the intrinsic (Bcl-2 and Mcl-1), extrinsic (TRAIL and Fas), p53-and endoplasmic reticulum (ER) stress-induced apoptotic pathways, as well as the necroptosis cell death pathway. In this review, we discuss the current knowledge of apoptosis and necroptosis pathways and how these are impaired in cancer cells. We focus on how miRNAs disrupt apoptosis and necroptosis, thereby critically contributing to malignancy. Understanding which and how miRNAs and their targets affect cell death pathways could open up novel therapeutic opportunities for cancer patients. Indeed, restoration of pro-apoptotic tumor suppressor miRNAs (apoptomiRs) or inhibition of oncogenic miRNAs (oncomiRs) represent strategies that are currently being trialed or are already applied as miRNA-based cancer therapies. Therefore, better understanding the cancer type-specific expression of apoptomiRs and oncomiRs and their underlying mechanisms in cell death pathways will not only advance our knowledge, but also continue to provide new opportunities to treat cancer.


Subject(s)
Apoptosis/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Necroptosis/genetics , Neoplasm Proteins/genetics , Neoplasms/genetics , Antagomirs/genetics , Antagomirs/metabolism , Antagomirs/therapeutic use , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Endoplasmic Reticulum Stress/genetics , Humans , MicroRNAs/agonists , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , Oligoribonucleotides/genetics , Oligoribonucleotides/metabolism , Oligoribonucleotides/therapeutic use , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , fas Receptor/genetics , fas Receptor/metabolism
11.
PLoS One ; 13(6): e0199258, 2018.
Article in English | MEDLINE | ID: mdl-29912984

ABSTRACT

Mutations in mitochondrial DNA are an important source of severe and incurable human diseases. The vast majority of these mutations are heteroplasmic, meaning that mutant and wild-type genomes are present simultaneously in the same cell. Only a very high proportion of mutant mitochondrial DNA (heteroplasmy level) leads to pathological consequences. We previously demonstrated that mitochondrial targeting of small RNAs designed to anneal with mutant mtDNA can decrease the heteroplasmy level by specific inhibition of mutant mtDNA replication, thus representing a potential therapy. We have also shown that 5S ribosomal RNA, partially imported into human mitochondria, can be used as a vector to deliver anti-replicative oligoribonucleotides into human mitochondria. So far, the efficiency of cellular expression of recombinant 5S rRNA molecules bearing therapeutic insertions remained very low. In the present study, we designed new versions of anti-replicative recombinant 5S rRNA targeting a large deletion in mitochondrial DNA which causes the KSS syndrome, analyzed their specific annealing to KSS mitochondrial DNA and demonstrated their import into mitochondria of cultured human cells. To obtain an increased level of the recombinant 5S rRNA stable expression, we created transmitochondrial cybrid cell line bearing a site for Flp-recombinase and used this system for the recombinase-mediated integration of genes coding for the anti-replicative recombinant 5S rRNAs into nuclear genome. We demonstrated that stable expression of anti-replicative 5S rRNA versions in human transmitochondrial cybrid cells can induce a shift in heteroplasmy level of KSS mutation in mtDNA. This shift was directly dependent on the level of the recombinant 5S rRNA expression and the sequence of the anti-replicative insertion. Quantification of mtDNA copy number in transfected cells revealed the absence of a non-specific effect on wild type mtDNA replication, indicating that the decreased proportion between mutant and wild type mtDNA molecules is not a consequence of a random repopulation of depleted pool of mtDNA genomes. The heteroplasmy change could be also modulated by cell growth conditions, namely increased by cells culturing in a carbohydrate-free medium, thus forcing them to use oxidative phosphorylation and providing a selective advantage for cells with improved respiration capacities. We discuss the advantages and limitations of this approach and propose further development of the anti-replicative strategy based on the RNA import into human mitochondria.


Subject(s)
DNA, Mitochondrial/genetics , Gene Transfer Techniques , Kearns-Sayre Syndrome/genetics , RNA, Ribosomal, 5S/genetics , Genetic Heterogeneity , Genetic Vectors/therapeutic use , Glucose/metabolism , Humans , Kearns-Sayre Syndrome/metabolism , Kearns-Sayre Syndrome/therapy , Mitochondria/genetics , Mitochondria/pathology , Mutation , Oligoribonucleotides/genetics , Oligoribonucleotides/therapeutic use , Transfection
12.
Int J Oncol ; 31(5): 1087-95, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17912434

ABSTRACT

We recently showed that therapy with 2'-5'-oligoadenylate (2-5A)-linked antisense against human telomerase RNA component (2-5A-anti-hTR) is a novel telomerase-targeting strategy against malignant gliomas. In this study, we investigated conventional chemotherapeutic agents and gamma-irradiation (IR) to determine whether they could augment the efficacy of 2-5A-anti-hTR against these tumors in vitro and in vivo. Treatment with 2-5A-anti-hTR inhibited the viability of U373-MG and U87-MG malignant glioma cells in a dose-dependent manner; the antitumor effect resulted from induction of apoptosis. Also, telomerase-positive astrocytes with oncogenic Ras were more sensitive to 2-5A-anti-hTR than were those without oncogenic Ras. In addition, we sought to determine the combined effect of 2-5A-anti-hTR with N, N'-bis (2-chloroethyl)-N-nitrosourea (BCNU), cisplatin (CDDP), paclitaxel (PTX), temozolomide (TMZ), or IR. When we administered the combination treatments on the same day, PTX and IR showed a greater combined effect with 2-5A-anti-hTR on both tumor cell lines than did BCNU, CDDP and TMZ. However, all of the combination regimens were synergistic when we first treated tumor cells with 2-5A-anti-hTR for 24 h and then exposed them to the conventional treatments. Apoptosis-inducing agents (CDDP and PTX) but not autophagy-inducing therapies (TMZ and IR) enhanced the incidence of apoptosis caused by 2-5A-anti-hTR. Lastly, we observed a combinatorial effect of 2-5A-anti-hTR and TMZ in vivo in subcutaneous U87-MG tumors in nude mice. Interestingly, treatment with TMZ increased the incidence of apoptosis in subcutaneous tumor cells treated with 2-5A-anti-hTR. These results suggest that 2-5A-anti-hTR is preferable in combination with established cancer therapies.


Subject(s)
Adenine Nucleotides/therapeutic use , Genetic Therapy/methods , Glioma/therapy , Oligonucleotides, Antisense/therapeutic use , Oligoribonucleotides/therapeutic use , RNA, Untranslated/antagonists & inhibitors , Telomerase/antagonists & inhibitors , Animals , Apoptosis , Astrocytes/enzymology , Autophagy , Cell Line, Tumor , Combined Modality Therapy , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Female , Glioma/pathology , Humans , Mice , RNA , RNA, Long Noncoding , RNA, Untranslated/genetics , Telomerase/genetics , Temozolomide
13.
Curr Opin Pharmacol ; 5(5): 502-7, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16081320

ABSTRACT

Although a prominent cause of upper and lower respiratory tract disease in infants and the elderly, clinical options for treatment of respiratory syncytial virus (RSV) infections remain limited. Historically, attempts to develop vaccines have been unsuccessful, and rapid viral mutation rates have stifled development of several small molecule-based antiviral agents. Thus, targeted approaches to block RSV replication, including humanized monoclonal antibodies and nucleic acid-based strategies (antisense and RNA interference), have emerged as potentially viable drug development options.


Subject(s)
Adenine Nucleotides/pharmacology , Adenine Nucleotides/therapeutic use , Oligoribonucleotides, Antisense/pharmacology , Oligoribonucleotides, Antisense/therapeutic use , Oligoribonucleotides/pharmacology , Oligoribonucleotides/therapeutic use , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Viruses/drug effects , Humans , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/genetics , Respiratory Syncytial Viruses/physiology , Virus Replication/drug effects
14.
Curr Opin Mol Ther ; 2(3): 318-24, 2000 Jun.
Article in English | MEDLINE | ID: mdl-11249626

ABSTRACT

In vitro selection constitutes a unique way to explore vast libraries (up to 10(14) to 10(15) different molecules) of randomly synthesized nucleic acids. Selected RNA or DNA sequences (aptamers) may help to understand the molecular interactions and processes of interest for human diseases. Aptamers can be chemically-modified for improved efficiency and use in complex biological media. Moreover, aptamers selected in vitro may retain their activity in vivo and thus offer novel perspectives for gene therapy and the design of new drugs.


Subject(s)
Oligonucleotides/therapeutic use , Animals , Anti-Bacterial Agents/metabolism , Base Sequence , Gene Expression , Gene Library , Genetic Therapy , Humans , Oligonucleotides/genetics , Oligonucleotides/metabolism , Oligoribonucleotides/genetics , Oligoribonucleotides/metabolism , Oligoribonucleotides/therapeutic use , Toxins, Biological/metabolism , Viral Proteins/genetics
16.
Patol Fiziol Eksp Ter ; (5): 39-42, 1991.
Article in Russian | MEDLINE | ID: mdl-1806892

ABSTRACT

The effect of enkad on the development of chloroquine retinopathy in rabbits was studied by electron microscopy and electrophysiological methods. It is shown that subconjunctival enkade injection inhibits the development of a dystrophic process in the retina induced by intravitreous chloroquine administration.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Chloroquine/antagonists & inhibitors , Oligoribonucleotides/therapeutic use , Retinal Degeneration/drug therapy , Ribonucleotides/therapeutic use , Animals , Female , Male , Rabbits , Retinal Degeneration/chemically induced
18.
Vascul Pharmacol ; 55(4): 92-105, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21802526

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNAs, that control diverse cellular functions by either promoting degradation or inhibition of target messenger RNA translation. An aberrant expression profile of miRNAs has been linked to human diseases, including cardiovascular dysfunction. This review summarizes the latest insights in the identification of vascular-specific miRNAs and their targets, as well as their roles and mechanisms in the vasculature. Furthermore, we discuss how manipulation of these miRNAs could represent a novel therapeutic approach in the treatment of vascular dysfunction.


Subject(s)
Cardiovascular Diseases/metabolism , Endothelium, Vascular/metabolism , MicroRNAs/physiology , Muscle, Smooth, Vascular/metabolism , Angiogenesis Modulating Agents/agonists , Angiogenesis Modulating Agents/antagonists & inhibitors , Angiogenesis Modulating Agents/metabolism , Animals , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/physiopathology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Gene Expression Regulation/drug effects , Humans , MicroRNAs/agonists , MicroRNAs/antagonists & inhibitors , Molecular Targeted Therapy , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiopathology , Oligoribonucleotides/therapeutic use , Oligoribonucleotides, Antisense/therapeutic use
19.
Vascul Pharmacol ; 55(4): 79-86, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21777698

ABSTRACT

The term angiogenesis derives from the Greek words 'angeio' meaning blood vessel, and 'genesis' meaning production or birth, together referring to the creation of blood vessels within the body. This term has been used to generally indicate the growth and remodeling process of the primitive vascular network into a complex network during pre-natal development. After birth, reparative angiogenesis is activated during wound healing and in response to ischemia, while pathological angiogenesis contributes to tumor growth and metastasis, arthritis and ocular diseases, such as diabetic retinopathy. MicroRNAs (miRNAs) are a class of endogenous, small, non-coding RNAs that control gene expression by acting on target mRNAs for promoting either their degradation or translational repression. There is increasing evidence that miRNAs play important roles in vascular development as well as in vascular diseases. In this review, we aim at describing the role of miRNAs in angiogenesis, focusing, in particular, on post-ischemic neovascularization. First, we will describe the regulation and the expression of miRNAs in endothelial cells. Then, we will analyze the role of miRNAs in reparative and pathological angiogenesis. Finally, we will discuss the innovative strategies available to inhibit the level of pathogenic anti-angiogenic miRNAs and to increase expression of therapeutic miRNAs.


Subject(s)
Angiogenesis Modulating Agents/metabolism , MicroRNAs/physiology , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic , Angiogenesis Modulating Agents/agonists , Angiogenesis Modulating Agents/antagonists & inhibitors , Angiogenesis Modulating Agents/therapeutic use , Animals , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Gene Expression Regulation/drug effects , Humans , MicroRNAs/agonists , MicroRNAs/antagonists & inhibitors , Molecular Targeted Therapy , Neovascularization, Pathologic/drug therapy , Neovascularization, Physiologic/drug effects , Oligoribonucleotides/therapeutic use , Oligoribonucleotides, Antisense/therapeutic use
20.
Vascul Pharmacol ; 55(4): 106-10, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21864716

ABSTRACT

Cardiometabolic diseases (CMD) (such as atherosclerosis, diabetes, and hypertension) are the primary cause of death and disability in the Western world. Although lifestyle programs and therapeutic approaches have significantly reduced the socio-economic burden of CMD, a large number of events still cannot be avoided (the so called residual risk). Recent developments in genetics and genomics provide a platform for investigating further this area with the aim of deepening our understanding of the atherosclerotic phenomena underlying CMD, for instance by providing better information on the type of subjects who would benefit the most from therapeutic interventions, or by discovering new genetic and metabolic derangements that may be targeted for the development of new interventions. MicroRNAs (miRNA) are short, non-coding RNAs that negatively regulate the expression of proteins by binding to specific sequences on the 3' region of target mRNAs. Bioinformatics analysis predicts that each miRNA may regulate hundreds of targets, suggesting that miRNAs may play roles in almost every biological pathway and process, including those of the cardiovascular system. Studies are beginning to unravel their fundamental importance in vessel biology. Here, we review recent advance regarding the involvement of miRNAs in arterial remodeling and atherosclerosis.


Subject(s)
Arteries/metabolism , Atherosclerosis/metabolism , MicroRNAs/physiology , Animals , Arteries/drug effects , Arteries/physiopathology , Atherosclerosis/drug therapy , Atherosclerosis/physiopathology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Gene Expression Regulation/drug effects , Humans , MicroRNAs/agonists , MicroRNAs/antagonists & inhibitors , Molecular Targeted Therapy , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiopathology , Oligoribonucleotides/therapeutic use , Oligoribonucleotides, Antisense/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL