Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Sci Adv ; 4(10): eaat3386, 2018 10.
Article in English | MEDLINE | ID: mdl-30345352

ABSTRACT

Antisense oligonucleotide (ASO) silencing of the expression of disease-associated genes is an attractive novel therapeutic approach, but treatments are limited by the ability to deliver ASOs to cells and tissues. Following systemic administration, ASOs preferentially accumulate in liver and kidney. Among the cell types refractory to ASO uptake is the pancreatic insulin-secreting ß-cell. Here, we show that conjugation of ASOs to a ligand of the glucagon-like peptide-1 receptor (GLP1R) can productively deliver ASO cargo to pancreatic ß-cells both in vitro and in vivo. Ligand-conjugated ASOs silenced target genes in pancreatic islets at doses that did not affect target gene expression in liver or other tissues, indicating enhanced tissue and cell type specificity. This finding has potential to broaden the use of ASO technology, opening up novel therapeutic opportunities, and presents an innovative approach for targeted delivery of ASOs to additional cell types.


Subject(s)
Drug Delivery Systems/methods , Glucagon-Like Peptide-1 Receptor/metabolism , Insulin-Secreting Cells/drug effects , Oligonucleotides, Antisense/administration & dosage , Animals , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Gene Silencing , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/genetics , HEK293 Cells , Humans , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/pharmacokinetics , RNA, Long Noncoding/genetics
2.
Diabetologia ; 54(4): 935-44, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21190014

ABSTRACT

AIMS/HYPOTHESIS: Insulin action is purportedly modulated by Drosophila tribbles homologue 3 (TRIB3), which in vitro prevents thymoma viral proto-oncogene (AKT) and peroxisome proliferator-activated receptor-γ (PPAR-γ) activation. However, the physiological impact of TRIB3 action in vivo remains controversial. METHODS: We investigated the role of TRIB3 in rats treated with either a control or Trib3 antisense oligonucleotide (ASO). Tissue-specific insulin sensitivity was assessed in vivo using a euglycaemic-hyperinsulinaemic clamp. A separate group was treated with the PPAR-γ antagonist bisphenol-A-diglycidyl ether (BADGE) to assess the role of PPAR-γ in mediating the response to Trib3 ASO. RESULTS: Trib3 ASO treatment specifically reduced Trib3 expression by 70% to 80% in liver and white adipose tissue. Fasting plasma glucose, insulin concentrations and basal rate of endogenous glucose production were unchanged. However, Trib3 ASO increased insulin-stimulated whole-body glucose uptake by ~50% during the euglycaemic-hyperinsulinaemic clamp. This was attributable to improved skeletal muscle glucose uptake. Despite the reduction of Trib3 expression, AKT2 activity was not increased. Trib3 ASO increased white adipose tissue mass by 70% and expression of Ppar-γ and its key target genes, raising the possibility that Trib3 ASO improves insulin sensitivity primarily in a PPAR-γ-dependent manner. Co-treatment with BADGE blunted the expansion of white adipose tissue and abrogated the insulin-sensitising effects of Trib3 ASO. Finally, Trib3 ASO also increased plasma HDL-cholesterol, a change that persisted with BADGE co-treatment. CONCLUSIONS/INTERPRETATION: These data suggest that TRIB3 inhibition improves insulin sensitivity in vivo primarily in a PPAR-γ-dependent manner and without any change in AKT2 activity.


Subject(s)
Insulin Resistance/physiology , PPAR gamma/metabolism , Protein Kinases/metabolism , Animals , Benzhydryl Compounds , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Epoxy Compounds/pharmacology , Glucose Clamp Technique , Immunoblotting , Insulin Resistance/genetics , Male , Oligonucleotides, Antisense/genetics , PPAR gamma/antagonists & inhibitors , PPAR gamma/genetics , Protein Kinases/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
3.
J Thromb Haemost ; 8(11): 2349-57, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20727068

ABSTRACT

Coagulation factor (F)XI was first described as a member of the contact pathway of coagulation. However, the 'classic' theory of the extrinsic and intrinsic pathway has been revised and FXI was found to be activated by thrombin and to play a role in sustained thrombin generation and fibrinolysis inhibition. Recent studies have pointed to a disproportionate role of FXI in thrombosis and hemostasis. The observations that human congenital FXI deficiency is generally accompanied by mild and injury-related bleeding, and that experimental, provoked bleeding in animals is unaffected by FXI deficiency or FXI inhibition, suggest that the FXI amplification pathway is less important for normal hemostasis in vivo. In contrast, elevated plasma levels of FXI may contribute to human thromboembolic disease and the antithrombotic efficacy of FXI inhibition has been demonstrated in numerous animal models of arterial, venous and cerebral thrombosis. Whether severe FXI deficiency in humans protects against thromboembolic events remains unclear, although some evidence exists that the occurrence of ischemic stroke or venous thrombosis is low in severely FXI-deficient patients. Because of its distinctive function in thrombosis and hemostasis, FXI is an attractive target for the treatment and prevention of thromboembolism. A novel strategy for FXI inhibition is the use of antisense technology which has been studied in various thrombosis and bleeding animal models. The results are promising and support the concept that targeting FXI might serve as a new, effective and potentially safer alternative for the treatment of thromboembolic disease in humans.


Subject(s)
Factor XI Deficiency/therapy , Factor XI/antagonists & inhibitors , Factor XI/metabolism , Fibrinolytic Agents/therapeutic use , Thrombosis/drug therapy , Animals , Blood Coagulation , Disease Models, Animal , Dose-Response Relationship, Drug , Hemostasis , Humans , Mice , Models, Biological , Oligonucleotides, Antisense/pharmacology , Reperfusion Injury , Thromboembolism/prevention & control , Thromboembolism/therapy , Warfarin/therapeutic use
4.
Neuroscience ; 154(3): 1077-87, 2008 Jun 26.
Article in English | MEDLINE | ID: mdl-18511207

ABSTRACT

Activation of the spinal phospholipase A(2) (PLA(2)) -cyclooxygenase (COX) -prostaglandin signaling pathway is widely implicated in nociceptive processing. Although the role of spinal COX isoforms in pain signal transmission has been extensively characterized, our knowledge of PLA(2) enzymes in this cascade is limited. Among all PLA(2) groups, cytosolic calcium-dependent PLA(2) group IVA (cPLA(2)IVA) appears to be the predominant PLA(2) enzyme in the spinal cord. In the present study we sought to (i) characterize anatomical and cellular distribution and localization of cPLA(2)IVA in dorsal horn of rat spinal cord, (ii) verify efficacy and selectivity of intrathecal (IT) delivery of an antisense oligonucleotide (AS) targeting rat cPLA(2)IVA mRNA on spinal expression of this enzyme, and (iii) examine the effect of down-regulation of spinal cPLA(2)IVA on peripheral tissue injury-induced pain behavior. Here we demonstrate that cPLA(2)IVA is constitutively expressed in rat spinal cord, predominantly in dorsal horn neurons and oligodendrocytes but not in astrocytes or microglia. Intrathecal injection of AS significantly down-regulated both protein and gene expression of cPLA(2)IVA in rat spinal cord, while control missense oligonucleotide (MS) had no effect. Immunocytochemistry confirmed that the reduction occurred in neurons and oligodendrocytes. cPLA(2)IVA AS did not alter expression of several other PLA(2) isoforms, such as secretory PLA(2) (groups IIA and V) and calcium-independent PLA(2) (group VI), indicating that the AS was specific for cPLA(2)IVA. This selective knockdown of spinal cPLA(2)IVA did not change acute nociception (i.e. paw withdrawal thresholds to acute thermal stimuli and intradermal formalin-induced first phase flinching), however, it significantly attenuated formalin-induced hyperalgesia (i.e. second phase flinching behavior), which reflects spinal sensitization. Thus the present findings suggest that cPLA(2)IVA may specifically participate in spinal nociceptive processing.


Subject(s)
Cytosol/enzymology , Formaldehyde , Hyperalgesia/prevention & control , Hyperalgesia/psychology , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/therapeutic use , Phospholipases A2/biosynthesis , Spinal Cord/enzymology , Animals , Behavior, Animal/drug effects , Blotting, Western , Cytosol/drug effects , Down-Regulation/drug effects , Hot Temperature , Hyperalgesia/chemically induced , Immunohistochemistry , Injections, Spinal , Male , Pain Measurement/drug effects , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/drug effects
5.
Gene Ther ; 11(22): 1665-72, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15470482

ABSTRACT

In an effort to improve the efficacy of antisense delivery, we evaluated polyethyleneimine (PEI, 2 kDa) alone or grafted with nonionic amphiphilic block copolymer Pluronic (P85) as a carrier for Ku86 antisense oligonucleotide (ASO) delivery. Ku86 is an abundant nuclear protein that plays an important role in nonhomologous DNA end joining and has implications in tumorigenesis and acquired drug resistance. Transfection of adherent and suspension cell lines with Ku86 ASOs complexed with P85-g-PEI (2 kDa) conjugates was associated with a specific decrease in Ku86 mRNA levels (EC50<75 nM and EC50<250 nM, respectively, n=3). More importantly, no requirement for reduced serum conditions was necessary during transfection. In contrast, whereas Ku86 ASOs complexed with PEI (2 kDa) alone were effective in decreasing Ku86 mRNA levels in adherent cell lines (EC50<75 nM, n=3), the formulation did not produce any detectable decrease in Ku86 mRNA levels in suspension cell lines. Transfection of adherent cell lines with 500 nM Ku86 ASOs formulated with P85-g-PEI (2 kDa) was associated with a specific decrease (<10% remaining of control) in Ku86 protein expression and a two-fold increased cell death after treatment with ionizing radiation (IR). In athymic nude mice bearing subcutaneous human HT29 colon adenocarcinoma xenografts, Ku86 ASO-P85-g-PEI (2 kDa) administration (15 mg/kg, subcutaneously) with a Q1D x 7 treatment schedule, when combined with a single dose of IR (6 Gy), caused a significant inhibition of HT29 tumor growth compared with mismatch- and naked antisense-pretreated control groups (time from 200 to 1000 mm3, 126.9 versus 84.18 and 87.76 days, P<0.005). A potentiation of the antitumor activity was observed in all mice treated with Ku86 ASO-P85-g-PEI (2 kDa) formulation; however, tumor growth inhibition was reversible upon treatment cessation. No morbidity/mortality or changes in histopathology were observed under this treatment regiment. Our results indicate that P85-g-PEI (2 kDa) conjugates may increase the efficacy of Ku86 ASO delivery in management of resistant malignancies, thus providing a rationale for their evaluation in cancer patients in combination with conventional anticancer therapies.


Subject(s)
Antigens, Nuclear/genetics , DNA-Binding Proteins/genetics , Genetic Therapy/methods , Neoplasms/therapy , Oligonucleotides, Antisense/administration & dosage , Transfection/methods , Animals , Cell Line, Tumor , Female , Gene Expression , Humans , Ku Autoantigen , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Poloxalene , Polyethyleneimine , Transplantation, Heterologous
6.
Br J Cancer ; 89(7): 1352-7, 2003 Oct 06.
Article in English | MEDLINE | ID: mdl-14520471

ABSTRACT

Advanced colon cancer is a malignancy with poor response to various treatment modalities including ionising radiation (IR) and chemotherapy. Both IR and chemotherapeutic agents have been shown to act by inducing apoptosis, a type of cell death antagonised by the Bcl-x(L) gene product. Since approximately 60% of human colon cancers express Bcl-x(L), it was the aim of this study to explore the potential of Bcl-x(L) antisense oligonucleotides as a novel radiosensitisation strategy. Caco-2 colon cancer cells were treated with Bcl-x(L) antisense oligonucleotides in combination with IR or cisplatin, and Bcl-x(L) protein expression, apoptosis, cell viability and clonogenic survival were examined. Bcl-x(L) antisense oligonucleotide specifically reduced the Bcl-x(L) protein level by almost 50% in Caco-2 cells. The decreased threshold for the induction of apoptosis resulted in a 300% increase of apoptosis after IR or cisplatin treatment and led to a 60% reduction of cell proliferation beyond response rates achieved with IR. These data suggest that Bcl-x(L) is an important factor contributing to the treatment resistance of human colon cancer. Specific reduction of Bcl-x(L) protein levels by antisense oligonucleotides qualifies as a promising therapeutic strategy for colon cancer that may help overcome resistance and improve clinical outcome in this malignancy.


Subject(s)
Apoptosis/radiation effects , Colorectal Neoplasms/radiotherapy , Oligonucleotides, Antisense/therapeutic use , Proto-Oncogene Proteins c-bcl-2/genetics , Radiation-Sensitizing Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Blotting, Western , Caco-2 Cells/radiation effects , Cell Division/drug effects , Cisplatin/therapeutic use , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Down-Regulation , Flow Cytometry , Humans , Proto-Oncogene Proteins c-bcl-2/metabolism , Radiation, Ionizing , Transfection , Tumor Stem Cell Assay , bcl-X Protein
7.
Br J Cancer ; 85(11): 1753-8, 2001 Nov 30.
Article in English | MEDLINE | ID: mdl-11742498

ABSTRACT

c-Raf is an essential component of the extracellular related kinase (ERK) signal transduction pathway. Immunohistochemical staining indicated that c-Raf was present in 49/53 ovarian adenocarcinomas investigated and high c-Raf expression correlated significantly with poor survival (P = 0.002). c-Raf protein was detected in 15 ovarian cancer cell lines. Antisense oligodeoxynucleotides (ODNs) (ISIS 5132 and ISIS 13650) reduced c-Raf protein levels and inhibited cell proliferation in vitro. Selectivity was demonstrated by the lack of effect of ISIS 5132 on A-Raf or ERK, while a random ODN produced only minor effects on growth and did not influence c-Raf expression. ISIS 5132 produced enhanced apoptosis and cells accumulated in S and G(2)/M phases of the cell cycle. In vivo, ISIS 5132 inhibited growth of the s.c. SKOV-3 xenograft while a mismatch ODN had no effect. These data indicate that high levels of c-Raf expression may be important in ovarian cancer and use of antisense ODNs targeted to c-Raf could provide a strategy for the treatment of this disease.


Subject(s)
DNA, Antisense/pharmacology , Ovarian Neoplasms/drug therapy , Proto-Oncogene Proteins c-raf/drug effects , Animals , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Division/drug effects , DNA, Antisense/therapeutic use , Dose-Response Relationship, Drug , Female , Humans , Immunohistochemistry , Mice , Mice, Nude , Neoplasm Transplantation , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins c-raf/biosynthesis , Proto-Oncogene Proteins c-raf/genetics , Survival Analysis , Time Factors , Xenograft Model Antitumor Assays
8.
Cancer Res ; 61(19): 7079-90, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11585739

ABSTRACT

Elevated focal adhesion kinase (FAK) expression in human tumor cells has been correlated with an increased cell invasion potential. In cell culture, studies with FAK-null fibroblasts have shown that FAK function is required for cell migration. To determine the role of elevated FAK expression in facilitating epidermal growth factor (EGF)-stimulated human adenocarcinoma (A549) cell motility, antisense oligonucleotides were used to reduce FAK protein expression >75%. Treatment of A549 cells with FAK antisense (ISIS 15421) but not a mismatched control (ISIS 17636) oligonucleotide resulted in reduced EGF-stimulated p130(Cas)-Src complex formation, c-Jun NH(2)-terminal kinase (JNK) activation, directed cell motility, and serum-stimulated cell invasion through Matrigel. Because residual FAK protein in ISIS 15421-treated A549 cells was highly phosphorylated at the Tyr-397/Src homology (SH)2 binding site, expression of the FAK COOH-terminal domain (FRNK) was also used as an inhibitor of FAK function. Adenoviral-mediated infection and expression of FRNK promoted FAK dephosphorylation at Tyr-397, resulted in reduced EGF-stimulated JNK as well as extracellular-regulated kinase 2 (ERK2) kinase activation, inhibited matrix metalloproteinase-9 (MMP-9) secretion, and potently blocked both random and EGF-stimulated A549 cell motility. Equivalent expression of a FRNK (S-1034) point-mutant that did not promote FAK dephosphorylation also did not affect EGF-stimulated signaling or cell motility. Dose-dependent reduction in EGF-stimulated A549 motility was observed with the PD98059 MEK1 inhibitor and the batimastat (BB-94) inhibitor of MMP activity, but not with the SB203580 inhibitor of p38 kinase. Finally, comparisons between normal, FAK-null, and FAK-reconstituted fibroblasts revealed that FAK enhanced EGF-stimulated JNK and ERK2 kinase activation that was required for cell motility. These data indicate that FAK functions as an important signaling platform to coordinate EGF-stimulated cell migration in human tumor cells and support a role for inhibitors of FAK expression or activity in the control of neoplastic cell invasion.


Subject(s)
Adenocarcinoma/enzymology , Cell Movement/physiology , Epidermal Growth Factor/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Adenocarcinoma/pathology , Cell Movement/drug effects , Enzyme Activation , Epidermal Growth Factor/pharmacology , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Invasiveness , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Phosphorylation , Protein-Tyrosine Kinases/biosynthesis , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/physiology , Tumor Cells, Cultured
9.
J Pharmacol Exp Ther ; 298(3): 934-40, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11504787

ABSTRACT

Phosphorothioate (P=S) antisense oligonucleotides (ASO) targeting the cell survival gene clusterin synergistically enhance castration- and chemotherapy-induced apoptosis in prostate cancer xenografts. This study compares efficacy, tissue half-lives, and toxicity of P=S clusterin ASO to third-generation backbone 2'-O-(2-methoxy)ethyl (2'MOE) ribose-modified clusterin ASO. Northern analysis quantified changes in clusterin mRNA levels in human PC-3 cells and tumors. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay measured effects of combined clusterin ASO plus paclitaxel on PC-3 cell growth. Athymic mice bearing PC-3 tumors were treated with paclitaxel plus either P=S clusterin ASO, 2'-MOE clusterin ASO, or mismatch control oligonucleotides for 28 days. Weekly body weights and serum parameters were measured to assess toxicity. Tissue half-life of P=S and 2'-MOE ASO in PC-3 tumors was assessed using capillary gel electrophoresis (CGE). Both 2'-MOE and P=S ASO decreased clusterin mRNA levels in a dose-dependent and sequence-specific manner. 2'-MOE ASO more potently suppressed clusterin mRNA (80 versus 40% at 500 nM) compared with P=S ASO. IC(50) of paclitaxel was equally reduced (50--75%) by both compounds. In vivo tissue half-life was significantly longer for 2'-MOE-modified ASO than for P=S ASO (5 versus 0.5 days). Using CGE, >90% of detected 2'-MOE ASO in tumor tissue was full length. Weekly administration of 2'-MOE clusterin ASO was equivalent to daily P=S clusterin ASO in enhancing paclitaxel efficacy in vivo. 2'-MOE-modified ASO potently suppressed clusterin expression and prolonged tissue half-lives with no additional side effects. These results support the use of 2'-MOE-modified ASO over conventional P=S ASO by potentially increasing potency and allowing longer dosing intervals in clinical trials.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Glycoproteins/genetics , Molecular Chaperones/genetics , Neoplasm Proteins/genetics , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents, Phytogenic/pharmacology , Blotting, Northern , Clusterin , Electrophoresis, Capillary , Glycoproteins/biosynthesis , Half-Life , Humans , Male , Mice , Molecular Chaperones/biosynthesis , Neoplasm Proteins/biosynthesis , Oligonucleotides, Antisense/pharmacokinetics , Paclitaxel/pharmacology , Prostatic Neoplasms/metabolism , RNA, Messenger/biosynthesis , Tetrazolium Salts , Thiazoles , Tumor Cells, Cultured
10.
Endocrinology ; 142(8): 3405-13, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11459784

ABSTRACT

Despite the importance of relaxin to normal parturition in various species and its potential as an etiological agent in preterm delivery in women, knowledge regarding the mechanisms by which relaxin alters cervical connective tissue is extremely limited. An established in vitro model for human pregnancy cervix, human lower uterine segment fibroblasts, was used to determine the effects of relaxin as well as those of progesterone on the expression of matrix metalloproteinases and tissue inhibitor of metalloproteinase-1. The results demonstrate that relaxin is a positive regulator of matrix metalloproteinase expression, as it stimulates the expression of procollagenase protein and mRNA levels, stimulates prostromelysin-1 protein and mRNA levels, and inhibits tissue inhibitor of metalloproteinase-1 protein expression. Stimulation of procollagenase and prostromelysin-1 expression by relaxin does not involve phorbol-12-myristate-13-acetate- sensitive PKCs. Relaxin-stimulated tyrosine phosphorylation of the putative receptor and inhibition by a receptor tyrosine kinase inhibitor suggest that the relaxin receptor is probably a tyrosine kinase receptor. Inhibition of c-Raf protein expression using an antisense oligonucleotide inhibits relaxin regulation of matrix metalloproteinase and tissue inhibitor of metalloproteinase-1, suggesting that a signaling pathway involving c-Raf kinase mediates relaxin action.


Subject(s)
Fibroblasts/enzymology , Matrix Metalloproteinases/metabolism , Protein-Tyrosine Kinases/physiology , Relaxin/pharmacology , Signal Transduction , Uterus/enzymology , Cells, Cultured , Collagenases/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Precursors/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Matrix Metalloproteinase Inhibitors , Metalloendopeptidases/metabolism , Phosphorylation/drug effects , Pregnancy , Progesterone/pharmacology , Protein Kinase C/physiology , Proto-Oncogene Proteins c-raf/physiology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Relaxin/physiology , Signal Transduction/physiology , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tyrosine/metabolism , Uterus/cytology , Uterus/drug effects , Uterus/metabolism
11.
Clin Cancer Res ; 7(5): 1214-20, 2001 May.
Article in English | MEDLINE | ID: mdl-11350886

ABSTRACT

Raf-1 is a serine/threonine kinase that functions as a critical effector of Ras-mediated signal transduction via the mitogen-activated protein kinase pathway. Constitutive activation of this pathway directly contributes to malignant transformation in many human tumors. A 20-base phosphorothioate oligonucleotide complementary to c-raf-1 mRNA (ISIS 5132; CGP 69846A) has been shown to specifically suppress Raf-1 expression both in vitro and in vivo. This Phase I trial, involving 22 patients with advanced cancer, was designed to evaluate the safety, feasibility, and maximum tolerated dose of ISIS 5132 administration as a weekly 24-h i.v. infusion. Pharmacokinetic analysis was performed, and c-raf-1 mRNA levels in peripheral blood mononuclear cells were assessed using quantitative reverse transcription-PCR. This trial defined a maximum tolerated dose of 24 mg/kg/week on this schedule. Two of four patients treated at 30 mg/kg/week had serious adverse events after the first dose of ISIS 5132, including acute hemolytic anemia and acute renal failure and anasarca. There were no major responses documented. Dose-dependent complement activation was demonstrated on this schedule, but not on previously evaluated schedules, of ISIS 5132 administration. In contrast to other trials of ISIS 5132, there appeared to be no consistent suppression of peripheral blood mononuclear cell c-raf-1 mRNA level on this schedule at any of the dose levels analyzed. These data suggest that the efficacy and toxicity profiles of antisense oligonucleotides may be highly dependent on the schedule of administration and support the analysis of the putative molecular target in the evaluation of novel therapeutics.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Oligodeoxyribonucleotides, Antisense/therapeutic use , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Thionucleotides/therapeutic use , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Blood Coagulation/drug effects , Complement System Proteins/metabolism , Drug Administration Schedule , Female , Humans , Infusions, Intravenous , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Neoplasm Staging , Neoplasms/metabolism , Oligodeoxyribonucleotides, Antisense/adverse effects , Oligodeoxyribonucleotides, Antisense/pharmacokinetics , Proto-Oncogene Proteins c-raf/genetics , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/blood , Thionucleotides/adverse effects , Thionucleotides/pharmacokinetics , Treatment Outcome
12.
Nucleic Acids Res ; 29(8): 1683-9, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11292840

ABSTRACT

2'-O-(2-methoxyethyl) (2'-MOE) RNA possesses favorable pharmocokinetic properties that make it a promising option for the design of oligonucleotide drugs. Telomerase is a ribonucleoprotein that is up-regulated in many types of cancer, but its potential as a target for chemotherapy awaits the development of potent and selective inhibitors. Here we report inhibition of human telomerase by 2'-MOE RNA oligomers that are complementary to the RNA template region. Fully complementary oligomers inhibited telomerase in a cell extract with IC(50) values of 5-10 nM at 37 degrees C. IC(50) values for mismatch-containing oligomers varied with length and phosphorothioate substitution. After introduction into DU 145 prostate cancer cells inhibition of telomerase activity persisted for up to 7 days, equivalent to six population doublings. Inside cells discrimination between complementary and mismatch-containing oligomers increased over time. Our results reveal two oligomers as especially promising candidates for initiation of in vivo preclinical trials and emphasize that conclusions regarding oligonucleotide efficacy and specificity in cell extracts do not necessarily offer accurate predictions of activity inside cells.


Subject(s)
Enzyme Inhibitors/metabolism , Oligonucleotides/chemistry , Oligonucleotides/metabolism , RNA, Antisense/chemistry , RNA, Antisense/metabolism , Telomerase/antagonists & inhibitors , Animals , Base Pair Mismatch/genetics , Base Pairing , Base Sequence , DNA, Recombinant/chemistry , DNA, Recombinant/genetics , DNA, Recombinant/metabolism , Enzyme Inhibitors/chemistry , Genetic Engineering , Humans , Inhibitory Concentration 50 , Mice , Mice, Nude , Neoplasm Transplantation , Oligonucleotides/genetics , Oligoribonucleotides , RNA/chemistry , RNA/genetics , RNA/metabolism , RNA, Antisense/genetics , Substrate Specificity , Telomerase/genetics , Telomerase/metabolism , Time Factors , Transfection , Tumor Cells, Cultured
13.
Mol Pharmacol ; 59(4): 867-74, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11259632

ABSTRACT

In the present study, rat cardiac myocytes were used as an in vitro ischemia/reperfusion injury model to delineate the role of c-Jun N-terminal kinase (JNK) 1 and JNK2 isoforms in ischemia/reoxygenation-induced apoptosis using an antisense approach. Exposure of rat cardiac myocytes to ischemia did not induce apoptosis as detected by staining with either acridine orange/ethidium bromide or annexin-V-fluorescein/propidium iodide. In contrast, a time-dependent increase in the number of apoptotic cells was noted after reoxygenation of ischemic myocytes, whereas the level of necrotic cells remained unaltered. Reoxygenation, but not ischemia alone, also caused a time-dependent increase in JNK activation that preceded apoptosis induction. Treatment of cardiac myocytes with antisense (AS) oligonucleotides that specifically targeted either JNK1 or JNK2 significantly reduced both mRNA and protein expression of the target isoform but had no effect on the expression of the alternate isoform. Pretreatment of cardiac myocytes with JNK1 AS, but not JNK2 AS, resulted in almost complete attenuation of reoxygenation-induced apoptosis. Furthermore, control oligonucleotides for JNK1 AS or JNK2 AS had no effect on JNK mRNA or protein expression or reoxygenation-induced apoptosis, indicating a sequence-specific mode of action. Additional studies revealed that apoptosis induced by other JNK-activating stimuli, including ceramide, heat shock, and UV irradiation, was partly suppressed after treatment with JNK1 AS but not JNK2 AS. These findings demonstrate that the JNK1 isoform plays a preferential role in apoptosis induced by ischemia/reoxygenation as well as diverse JNK-activating cellular stresses.


Subject(s)
Apoptosis/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Myocardial Ischemia/metabolism , Myocardium/metabolism , Animals , Apoptosis/radiation effects , Cell Hypoxia/drug effects , Cells, Cultured , Enzyme Induction/physiology , Heat-Shock Response/drug effects , Isoenzymes/antagonists & inhibitors , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinase 9 , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Myocardium/cytology , Oligonucleotides, Antisense/pharmacology , Oxygen/pharmacology , Phosphorylation/drug effects , Phosphorylation/radiation effects , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Ultraviolet Rays
14.
Br J Haematol ; 112(3): 706-13, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11260076

ABSTRACT

Bcl-XL, a member of the Bcl-2-related anti-apoptosis protein family, antagonizes a diverse range of apoptosis-inducing stimuli by preventing mitochondrial permeability transition, release of apoptogenic factors including cytochrome C, and caspase activation. We have tested the hypothesis that the susceptibility of Bcl-XL-expressing leukaemic cells to apoptosis induced by VP16 (etoposide) can be enhanced by pharmacological downregulation of Bcl-XL in vivo. Two subcutaneous xenograft models of B-cell leukaemia-employing SEMK-2 and BV173 cell lines were established in severe combined immunodeficient/non-obese diabetic mice followed by 14 d of continuous subcutaneous administration of Bcl-XL-specific second generation oligonucleotides ISIS 16009 or ISIS 15999. Tumours were disaggregated, enabling investigation of Bcl-XL expression and apoptosis susceptibility at single-cell resolution using cytofluorimetry. Marked sequence-specific reduction of Bcl-XL was associated with sequence-specific enhancement of VP16-induced mitochondrial permeability transition, caspase-3 activation and loss of membrane asymmetry. A negative correlation between Bcl-XL expression and apoptosis susceptibility was observed, together with a positive correlation with respect to a reduced redox state. Bcl-XL downregulation reduces the threshold for VP16-induced apoptosis by potentiating mitochondrial dysfunction and its sequelae, and therefore presents a novel therapeutic strategy for reversing chemoresistance.


Subject(s)
Drug Resistance, Neoplasm/genetics , Genetic Therapy/methods , Oligonucleotides, Antisense/administration & dosage , Proto-Oncogene Proteins c-bcl-2/genetics , Severe Combined Immunodeficiency/drug therapy , Animals , Apoptosis/drug effects , Caspase 3 , Caspases/metabolism , Combined Modality Therapy , Enzyme Activation , Etoposide/pharmacology , Flow Cytometry/methods , Humans , Mice , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/metabolism , Transplantation, Heterologous , Tumor Cells, Cultured , bcl-X Protein
15.
Methods ; 23(2): 191-8, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11181038

ABSTRACT

In the past decade antisense oligonucleotides (ASOs) have proven to be a useful tool for dissection of gene function in molecular cell biology (Koller, E., Gaarde, W. A., and Monia, B. P. (2000) Trends Pharm. Sci., 21, 142-148), and validation of gene targets in animal models (Crooke, S. T. (1998) Biotechnol. Gen. Eng. Rev. 15, 121-157), as well as a means for therapeutic treatment of human diseases (Bennett, C. F. (1999) Exp. Opin. Invest. Drugs 8, 237-253). An important step toward usage of ASOs in the described applications is identification of an active ASO. This article describes the underlying basis and means for achieving this goal in cell culture.


Subject(s)
Genetic Techniques , Oligonucleotides, Antisense/metabolism , Animals , Blotting, Northern , Cell Line , Cells, Cultured , Humans , Oligonucleotide Array Sequence Analysis , RNA/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ribonuclease H/metabolism , Ribonucleases/metabolism
16.
Am J Respir Cell Mol Biol ; 24(2): 116-22, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11159044

ABSTRACT

The unique role of interleukin (IL)-5 in eosinophil production, activation, and localization makes this cytokine a prime target for therapeutic intervention in diseases characterized by a selective blood and tissue eosinophilia. In an attempt to block the effects of IL-5 on eosinophils, a strategy was developed to suppress the expression of the IL-5 receptor alpha chain (IL-5Ralpha) by antisense oligonucleotides (ASOs). IL-5Ralpha ASOs were identified which selectively and specifically suppress the expression of messenger RNA and proteins of both the membrane and the soluble form of the receptor in constitutively IL-5R-expressing murine BCL-1 cells in vitro. Moreover, these IL-5Ralpha-specific ASOs were able to selectively inhibit the IL-5-induced eosinopoesis from murine fetal liver and bone marrow cells in vitro, suggesting that these molecules may affect the development of IL-5-mediated eosinophilia in vivo. Indeed, intravenous administration of IL-5Ralpha-specific ASOs not only suppressed the bone-marrow and blood eosinophilia in mice after short-term treatment with recombinant murine IL-5 but also inhibited the development of blood and tissue eosinophilia in a ragweed-induced allergic peritonitis model. Thus, blocking the expression of IL-5Ralpha on eosinophil using ASOs may have therapeutic benefits in eosinophilic diseases such as asthma.


Subject(s)
Eosinophilia/prevention & control , Eosinophils/metabolism , Interleukin-5/pharmacology , Oligonucleotides, Antisense/pharmacology , Receptors, Interleukin/antagonists & inhibitors , Animals , Blotting, Northern , Blotting, Western , Bone Marrow/drug effects , Bone Marrow/metabolism , DNA Primers/chemistry , Eosinophilia/metabolism , Female , Humans , In Vitro Techniques , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Peritonitis/genetics , Peritonitis/pathology , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis , Receptors, Interleukin/metabolism , Receptors, Interleukin-5 , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Transfection , Tumor Cells, Cultured
17.
Antisense Nucleic Acid Drug Dev ; 10(5): 347-57, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11079574

ABSTRACT

Binding of human interleukin-5 (HuIL-5) to its membrane-anchored receptor (IL-5R) triggers multiple signaling pathways, cellular proliferation, and maturational responses, as well as protection from apoptosis. In contrast, soluble forms of the HuIL-5R have been shown to inhibit IL-5 signaling and, therefore, may represent naturally occurring negative regulators of IL-5 function. Because of the central role of IL-5 in promoting eosinophilia and airway hyperresponsiveness in animal models of asthma, antisense oligonucleotides specific either for the membrane form alone or for sequences shared between both the membrane and soluble forms of the HuIL-5Ralpha ligand binding chain were designed. The activities of these oligonucleotides were characterized in IL-5R-expressing erythroleukemic TF-1 cells. Herein we report that an antisense oligonucleotide targeted to a sequence unique to the alternatively spliced membrane-bound form of the HuIL-5Ralpha chain has been developed that selectively inhibits membrane, but not soluble, mRNA isoform expression. Both this membrane-specific oligonucleotide and an antisense oligonucleotide targeted to sequence common to both membrane and soluble isoforms were found to potently suppress cell surface IL-5Ralpha levels and IL-5-mediated cell survival by inducing apoptosis similar to IL-5 withdrawal. Thus, these oligonucleotides represent unique genetic agents with therapeutic potential for diseases with an eosinophilic component.


Subject(s)
Apoptosis , Membrane Proteins/biosynthesis , Oligonucleotides, Antisense/genetics , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism , Alternative Splicing/genetics , Apoptosis/drug effects , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Interleukin-5/pharmacology , Kinetics , Leukemia, Erythroblastic, Acute/genetics , Leukemia, Erythroblastic, Acute/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phosphorylation , Protein Isoforms/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Interleukin/biosynthesis , Receptors, Interleukin-5 , Signal Transduction/drug effects , Solubility , Substrate Specificity , Transfection , Tumor Cells, Cultured
19.
Transplantation ; 70(4): 656-61, 2000 Aug 27.
Article in English | MEDLINE | ID: mdl-10972225

ABSTRACT

BACKGROUND: C-raf is a well-characterized serine/ threonine (Ser/Thr) protein kinase that is involved in the transduction of multiple signals of T cells. We demonstrate that the inhibition of C-raf mRNA expression prolongs heart allograft survival. METHODS: Three 20-mer C-raf antisense oligonucleotides, each with identical sequences, were synthesized with different chemical modifications: one as a uniform phosphorothioate oligodeoxynucleotide (PS oligo), a second with a PS backbone and 2'-methoxyethyl (ME) substitutions at the 2'-sugar positions in the first and last five nucleotides, and a third with a mixed PS and phosphodiester (PD) backbone and ME modifications on the first and last five nucleotides. RESULTS: Both ME-modified C-raf antisense oligos were at least 5-fold more effective than the PS C-raf antisense oligo in blocking C-raf mRNA expression in two cell lines. Similarly, each of the ME C-raf antisense oligos produced better heart allograft survival rates than did PS C-raf oligo. Furthermore, although the combination of PS C-raf antisense oligo with sirolimus (SRL) acted synergistically to extend heart allograft survival, the effect was potentiated by either of the ME-modified oligos. CONCLUSIONS: C-raf inhibition extends heart allograft survival, and ME-modification potentiates antisense activity.


Subject(s)
Gene Expression Regulation/drug effects , Graft Survival/genetics , Heart Transplantation/physiology , Oligodeoxyribonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogenes , Animals , Base Sequence , Cell Line , Graft Survival/drug effects , Heart Transplantation/immunology , Humans , Mice , Rats , Rats, Inbred ACI , Rats, Inbred Lew , Thionucleotides , Transplantation, Homologous
20.
Mol Pharmacol ; 58(2): 380-7, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10908306

ABSTRACT

Expression of the interleukin-5 receptor-alpha (IL-5Ralpha) chain is thought to play an important role in the pathogenesis of asthma and other eosinophilic diseases. With antisense oligonucleotides (ASOs) chemically modified to provide increased hybridization affinity for RNA but that do not support RNase H-mediated cleavage (2'-O-methoxyethyl-modified ASOs), we show that constitutive splicing of murine IL-5Ralpha mRNA can be modulated in cells such that individual exons may be selectively deleted from mature transcripts. Specific deletion of individual exons and redirection of alternative splicing of the IL-5Ralpha mRNA have been achieved with this approach, by targeting 3'-splice sites or exon sequences immediately downstream of an alternative splice site. ASO targeting with these strategies resulted in inhibition of mRNA and protein levels of the membrane IL-5Ralpha isoform capable of signaling IL-5-mediated growth and antiapoptotic signals to eosinophils. Membrane isoform IL-5Ralpha inhibition was coupled with an increase in expression of mRNA for the alternatively spliced soluble isoform, which binds IL-5 extracellularly and may block its function. These observations suggest the potential general therapeutic use of an antisense approach to increase expression of variant RNA transcripts and to thereby produce proteins devoid of specific functional domains that may impact disease processes, as well as its specific utility for modulating expression of a key cytokine receptor implicated in allergic inflammation.


Subject(s)
Alternative Splicing/drug effects , Gene Expression Regulation/drug effects , Oligonucleotides, Antisense/pharmacology , RNA Precursors/metabolism , Receptors, Interleukin/genetics , Animals , Base Sequence , Exons/genetics , Gene Deletion , Mice , Molecular Sequence Data , Protein Isoforms , RNA Precursors/drug effects , Receptors, Interleukin/metabolism , Receptors, Interleukin-5 , Ribonuclease H/metabolism , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...