Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Gene Ther ; 13(24): 1724-30, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16886001

ABSTRACT

Depsipeptide, a histone deacetylase (HDAC) inhibitor, kills tumor cells much more effectively than normal cells, and can produce significant antitumor activity in human cancer patients. Depsipeptide also increases the expression of lipoplex-delivered genes in cultured tumor cells, as well as following direct intra-tumoral injection. We now show that co-intravenous (i.v.) injection of depsipeptide with polyethylenimine (PEI):DNA complexes significantly increases the expression of PEI-delivered genes in normal, as well as in tumor-bearing mice. At the tissue level, depsipeptide-mediated enhancement of gene expression was selectively targeted to the lung, liver and spleen. At the cellular level, depsipeptide significantly increased the expression of the i.v., PEI co-delivered wild-type human p53 gene in metastatic breast cancer cells, but not in adjacent normal cells. Thus, the ability of depsipeptide to enhance the expression of systemically delivered genes is selectively targeted at both the tissue and cellular levels, without requiring the use of ligand- or promoter-based approaches. Analyzing HDAC-based targeting of gene expression may identify host genes that control the expression of systemically delivered genes.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Depsipeptides/therapeutic use , Genetic Therapy/methods , Histone Deacetylase Inhibitors , Neoplasms/therapy , Transfection/methods , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Cell Line, Tumor , Colorimetry , Combined Modality Therapy , Female , Gene Expression/drug effects , Gene Targeting , Genes, p53 , Immunohistochemistry/methods , Injections, Intravenous , Liver/metabolism , Luciferases/analysis , Luciferases/genetics , Lung/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Neoplasms/drug therapy , Spleen/metabolism
2.
Cancer Gene Ther ; 13(3): 306-17, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16138118

ABSTRACT

Intravenous gene delivery using liposome-DNA complexes (LDC) has previously been shown to elicit antitumor activity, but only in rodent tumor models. Therefore, we conducted a study to determine in a large animal spontaneous tumor model whether intravenous infusions of LDC could target gene expression to cutaneous tumor tissues and whether repeated treatments had an effect on tumor growth or angiogenesis. A total of 13 dogs with cutaneous soft tissue sarcomas were enrolled in the study and were randomized to receive a series of 6 weekly infusions of LDC containing either canine endostatin DNA or DNA encoding an irrelevant gene (luciferase). Serial tumor biopsies were obtained to assess transgene expression, tumor microvessel density (MVD), and intratumoral leukocyte inflammatory responses. We found that intravenous infusion of LDC did not result in detectable gene expression in cutaneous tumor tissues. However, two of 13 treated dogs had objective tumor responses and eight dogs had stable disease during the treatment period. In addition, a significant decrease in tumor MVD was noted in six of 12 treated dogs at the completion of six treatments. These results suggest that intravenous infusions of LDC may elicit nonspecific antitumor activity and inhibit tumor angiogenesis.


Subject(s)
DNA/administration & dosage , Dog Diseases/prevention & control , Endostatins/genetics , Neovascularization, Pathologic/veterinary , Sarcoma/veterinary , Skin Neoplasms/veterinary , Animals , Dog Diseases/metabolism , Dogs , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Fibrosarcoma/blood supply , Fibrosarcoma/therapy , Fibrosarcoma/veterinary , Genetic Vectors , Infusions, Intravenous , Liposomes/administration & dosage , Luciferases/genetics , Luciferases/metabolism , Mice , Neovascularization, Pathologic/metabolism , Sarcoma/blood supply , Sarcoma/metabolism , Skin Neoplasms/blood supply , Skin Neoplasms/metabolism , Spleen/metabolism , Spleen/pathology , Transgenes/physiology , Vascular Endothelial Growth Factor A/metabolism
3.
J Exp Med ; 194(5): 669-76, 2001 Sep 03.
Article in English | MEDLINE | ID: mdl-11535634

ABSTRACT

Multiple sclerosis (MS) is a demyelinating disease of the central nervous system (CNS) characterized by plaques of infiltrating CD4(+) and CD8(+) T cells. Studies of MS and experimental autoimmune encephalomyelitis (EAE), an animal model of MS, focus on the contribution of CD4(+) myelin-specific T cells. The role of CD8(+) myelin-specific T cells in mediating EAE or MS has not been described previously. Here, we demonstrate that myelin-specific CD8(+) T cells induce severe CNS autoimmunity in mice. The pathology and clinical symptoms in CD8(+) T cell-mediated CNS autoimmunity demonstrate similarities to MS not seen in myelin-specific CD4(+) T cell-mediated EAE. These data suggest that myelin-specific CD8(+) T cells could function as effector cells in the pathogenesis of MS.


Subject(s)
Brain/immunology , CD8-Positive T-Lymphocytes/immunology , Multiple Sclerosis/immunology , Myelin Basic Protein/immunology , Spinal Cord/immunology , Adoptive Transfer , Animals , Autoimmunity , Brain/pathology , Brain/physiopathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Clone Cells , Disease Models, Animal , Mice , Mice, Inbred C3H , Mice, Inbred MRL lpr , Mice, SCID , Multiple Sclerosis/pathology , Multiple Sclerosis/physiopathology , Spinal Cord/pathology , Spinal Cord/physiopathology , T-Lymphocytes/immunology , Time Factors
4.
Mol Ther ; 3(5 Pt 1): 734-45, 2001 May.
Article in English | MEDLINE | ID: mdl-11356078

ABSTRACT

One of the obstacles to successful lung gene transfer is effective delivery of vector to lung, particularly injured or diseased lung. We have previously demonstrated that intratracheal instillation of perfluorochemical (PFC) liquids along with instillation of recombinant adenovirus and adeno-associated virus vectors, or with cationic liposome vectors, increased total lung gene expression and enhanced distribution of gene expression throughout the lung. To further explore the potential benefits of PFC liquid use, we evaluated the effect of PFC liquid instillation on several other aspects of adenovirus-mediated gene expression in lung. Use of PFC liquid resulted in earlier detection of gene expression and allowed the use of less vector to achieve expression comparable to that observed with the use of higher amounts of vector alone. Using PFC liquid also enhanced gene expression in a rodent model of acute lung injury. PFC liquid did cause a transient inflammation when instilled into normal lungs but did not cause any additional inflammation when instilled alone or with adenovirus vector into acutely injured lungs. Thus, PFC liquid may be a useful adjunct for clinical lung gene transfer, particularly for injured or diseased lungs.


Subject(s)
Fluorocarbon Polymers/chemistry , Gene Transfer Techniques , Genetic Vectors , Lung Injury , Lung/metabolism , Adenoviridae/genetics , Animals , Bronchoalveolar Lavage Fluid , Fluorocarbons/chemistry , Furans/chemistry , Humans , Immunohistochemistry , In Situ Hybridization , Interleukin-1/biosynthesis , Interleukin-6/biosynthesis , Male , Rats , Rats, Sprague-Dawley , Time Factors , Transgenes , Tumor Necrosis Factor-alpha/biosynthesis , beta-Galactosidase/metabolism
5.
J Invest Dermatol ; 116(1): 131-5, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11168808

ABSTRACT

Transfection of the skin by local gene delivery, as well as widespread transfection of systemic tissues following intravenous injection of cationic liposome/DNA complexes have been reported. Here, we show that surgically wounded mouse skin can be transfected either by local injection of DNA alone or by intravenous injection of optimized cationic liposome/DNA complexes; however, direct cutaneous injection produces much higher levels of gene expression in the skin, which is targeted to dermal and subdermal layers. High levels of chloramphenicol acetyltransferase activity were present from 3 h to 2 wk following direct injection of a gene expression plasmid into wounded skin and were maintained at detectable levels up to 8 wk after injection. Expression of transferred chloramphenicol acetyltransferase as well as beta-GAL genes was localized to fibroblasts, macrophages, and adipocytes as determined by histochemistry and immunohistochemistry. Further- more, local injection of a human granulocyte- colony-stimulating factor gene expression plasmid produced high levels of the biologically relevant human granulocyte-colony-stimulating factor protein in wounded mouse skin. This efficient and simple method of site-specific gene transfer into wounds may lead to the development of cutaneous gene therapy directed against disorders of abnormal cutaneous wound healing.


Subject(s)
Plasmids/administration & dosage , Wound Healing/genetics , Wounds and Injuries/genetics , Animals , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , Cytomegalovirus/genetics , DNA, Viral/analysis , Dose-Response Relationship, Drug , Female , Gene Expression , Granulocyte Colony-Stimulating Factor/genetics , Injections , Mice , Mice, Inbred ICR , Time Factors , Transfection
6.
Radiat Res ; 155(1 Pt 1): 2-14, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11121210

ABSTRACT

Radiation of the esophagus of C3H/HeNsd mice with 35 or 37 Gy of 6 MV X rays induces significantly increased RNA transcription for interleukin 1 (Il1), tumor necrosis factor alpha (Tnf), interferon gamma inducing factor (Ifngr), and interferon gamma (Ifng). These elevations are associated with DNA damage that is detectable by a comet assay of explanted esophageal cells, apoptosis of the esophageal basal lining layer cells in situ, and micro-ulceration leading to dehydration and death. The histopathology and time sequence of events are comparable to the esophagitis in humans that is associated with chemoradiotherapy of non-small cell lung carcinoma (NSCLC). Intraesophageal injection of clinical-grade manganese superoxide dismutase-plasmid/liposome (SOD2-PL) 24 h prior to irradiation produced an increase in SOD2 biochemical activity in explanted esophagus. An equivalent therapeutic plasmid weight of 10 microgram ALP plasmid in the same 500 microliter of liposomes, correlated to around 52-60% of alkaline phosphatase-positive cells in the squamous layer of the esophagus at 24 h. Administration of SOD2-PL prior to irradiation mediated a significant decrease in induction of cytokine mRNA by radiation and decreased apoptosis of squamous lining cells, micro-ulceration, and esophagitis. Groups of mice receiving 35 or 37 Gy esophageal irradiation by a technique protecting the lungs and treating only the central mediastinal area were followed to assess the long-term effects of radiation. SOD2-PL-treated irradiated mice demonstrated a significant decrease in esophageal wall thickness at day 100 compared to irradiated controls. Mice with orthotopic thoracic tumors composed of 32D-v-abl cells that received intraesophageal SOD2-PL treatment showed transgenic mRNA in the esophagus at 24 h, but no detectable human SOD2 transgene mRNA in explanted tumors by nested RT-PCR. These data provide support for translation of this strategy of SOD2-PL gene therapy to studies leading to a clinical trial in fractionated irradiation to decrease the acute and chronic side effects of radiation-induced damage to the esophagus.


Subject(s)
Cytokines/biosynthesis , Esophageal Stenosis/prevention & control , Esophagitis/prevention & control , Genetic Therapy/methods , Radiation Injuries/prevention & control , Radiation Protection/methods , Superoxide Dismutase/genetics , Animals , Apoptosis/radiation effects , Cytokines/genetics , Esophageal Stenosis/ethnology , Esophageal Stenosis/metabolism , Esophagitis/etiology , Esophagitis/metabolism , Female , Gene Expression , Humans , Liposomes , Male , Mediastinal Neoplasms/genetics , Mediastinal Neoplasms/metabolism , Mice , Mice, Inbred C3H , Plasmids , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Radiation Injuries/ethnology , Radiation Injuries/metabolism , Radiation Tolerance/genetics , Reverse Transcriptase Polymerase Chain Reaction , Superoxide Dismutase/metabolism , Transgenes
7.
J Immunol ; 165(6): 3418-22, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10975861

ABSTRACT

Allergen-induced asthma is characterized by chronic pulmonary inflammation, reversible bronchoconstriction, and airway hyperreactivity to provocative stimuli. Multiple CC-chemokines, which are produced by pulmonary tissue in response to local allergen challenge of asthmatic patients or experimentally sensitized rodents, chemoattract leukocytes from the circulation into the lung parenchyma and airway, and may also modify nonchemotactic function. To determine the therapeutic potential of local intrapulmonary CC-chemokine blockade to modify asthma, a recombinant poxvirus-derived viral CC-chemokine inhibitor protein (vCCI), which binds with high affinity to rodent and human CC-chemokines in vitro and neutralizes their biological activity, was administered by the intranasal route. Administration of vCCI to the respiratory tract resulted in dramatically improved pulmonary physiological function and decreased inflammation of the airway and the lung parenchyma. In contrast, vCCI had no significant effect on the circulating levels of total or allergen-specific IgE, allergen-specific cytokine production by peripheral lymph node T cells, or peritoneal inflammation after local allergen challenge, indicating that vCCI did not alter systemic Ag-specific immunity or chemoattraction at extrapulmonary sites. Together, these findings emphasize the importance of intrapulmonary CC-chemokines in the pathogenesis of asthma, and the therapeutic potential of generic and local CC-chemokine blockade for this and other chronic diseases in which CC-chemokines are locally produced.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Bronchial Hyperreactivity/pathology , Bronchial Hyperreactivity/prevention & control , Chemokines, CC/antagonists & inhibitors , Cowpox virus/immunology , Respiratory Hypersensitivity/pathology , Respiratory Hypersensitivity/prevention & control , Viral Proteins/administration & dosage , Administration, Intranasal , Animals , Anti-Asthmatic Agents/administration & dosage , Bronchial Hyperreactivity/immunology , Cowpox virus/genetics , Disease Models, Animal , Female , Humans , Immunity, Cellular/drug effects , Mice , Mice, Inbred BALB C , Ovalbumin/administration & dosage , Ovalbumin/immunology , Recombinant Fusion Proteins/administration & dosage , Respiratory Hypersensitivity/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Viral Proteins/genetics , Virulence Factors
8.
Int J Cancer ; 90(3): 128-37, 2000 Jun 20.
Article in English | MEDLINE | ID: mdl-10900424

ABSTRACT

Esophagitis is a major limiting factor in the treatment of lung cancer by radiation alone or in combination with chemotherapy. We have previously demonstrated that intraesophageal injection of manganese superoxide dismutase-plasmid/liposome (MnSOD-PL) complex into C3H/HeNsd mice blocks irradiation-induced esophagitis. To determine whether the human esophagus can be similarly transfected, normal human esophageal sections obtained from the margins of esophagectomy specimens from esophageal cancer patients were transfected in vitro with alkaline phosphatase (AlkP)-PL complex and stained for AlkP activity, and the percent of cells expressing AlkP was calculated. At 24 hr after transfection with 20 or 200 microgram of AlkP-PL complex, 55.0% and 85.8% of esophageal epithelial cells expressed detectable AlkP, respectively. Other sections transfected with MnSOD-PL complex showed transgene mRNA by nested reverse transcriptase-polymerase chain reaction (RT-PCR) assay and increased MnSOD biochemical activity for at least 96 hr after transfection. Irradiated MnSOD-PL complex-transfected sections demonstrated a significantly decreased percentage of apoptotic cells when compared to irradiated control sections. Following 1,000 cGy, MnSOD-PL-treated samples showed 7.5 +/- 2.8% and 33.3 +/- 7.3% apoptotic cells at 24 and 48 hr compared to 53.6 +/- 6.9% and 59.0 +/- 13.8% for nontransfected controls (P < 0.0001 and P < 0.1175). After 2,000 cGy, results at 24 and 48 hr were 25.0 +/- 7.6% and 66.9 +/- 4.9% for MnSOD-transfected sections compared to 65.6 +/- 4.3% and 90.0 +/- 4.1% for control sections (P < 0.0001 and P = 0.0353), respectively. Thus, human esophageal sections can be transfected with MnSOD-PL complex in vitro and thereby protected against ionizing irradiation-induced apoptosis. Int. J. Cancer (Radiat. Oncol. Invest.) 90, 128-137 (2000).


Subject(s)
Apoptosis/radiation effects , Esophagus/radiation effects , Genetic Therapy , Radiation Protection , Superoxide Dismutase/genetics , Alkaline Phosphatase/genetics , Animals , Esophagus/enzymology , Humans , Liposomes , Mice , Mice, Inbred C3H , Plasmids , Transgenes
9.
J Biol Chem ; 275(39): 30408-16, 2000 Sep 29.
Article in English | MEDLINE | ID: mdl-10856307

ABSTRACT

To date, no gene transfer vector has produced prolonged gene expression following a single intravenous injection and then efficiently re-expressed the delivered gene following repeated systemic injection into immunocompetent hosts. To overcome these limitations, a gene therapy regimen using non-replicating Epstein-Barr virus (EBV)-based expression plasmids was developed. One plasmid contains the FR (EBV family of repeats) sequence and the expressed gene. The other encodes Epstein-Barr nuclear antigen 1 (EBNA-1), but lacks FR. Although unable to replicate in mice, intravenous co-injection of EBV-based plasmids in cationic liposome-DNA complexes (CLDCs) substantially prolonged luciferase gene expression. The use of a two-vector system limited host exposure to the EBNA-1 gene product. Furthermore, this EBV-based vector system could be intravenously re-injected multiple times into immunocompetent mice without loss of transfection efficiency. Use of this vector system significantly improved the therapeutic efficacy of the biologically important human granulocyte colony-stimulating factor gene. Delivery of the human granulocyte colony-stimulating factor gene in EBV-based plasmids increased circulating white blood counts for at least 2 months following a single CLDC-based intravenous co-injection. Conversely, white blood counts were never elevated following injection of CLDCs lacking EBV-derived elements. Thus, this EBV-based plasmid vector system both markedly prolongs gene expression at therapeutic levels and efficiently and repeatedly re-transfects immunocompetent hosts. These properties of EBV-based plasmid vectors appear to be due, at least in part, to the documented abilities of the EBNA-1 protein both to retain FR-containing DNA intracellularly and within the nucleus and to block anti-EBNA-1 cytotoxic T cell responses.


Subject(s)
DNA, Viral/genetics , Genetic Therapy/methods , Granulocyte Colony-Stimulating Factor/administration & dosage , Herpesvirus 4, Human/genetics , Plasmids/genetics , Animals , Drug Carriers , Epstein-Barr Virus Nuclear Antigens/genetics , Female , Gene Expression , Genes, Reporter , Granulocyte Colony-Stimulating Factor/biosynthesis , Granulocyte Colony-Stimulating Factor/genetics , Herpesvirus 4, Human/growth & development , Humans , Injections, Intravenous , Liposomes , Luciferases/biosynthesis , Luciferases/genetics , Mice , Mice, Inbred Strains , Repetitive Sequences, Nucleic Acid , Virus Replication
10.
Nat Biotechnol ; 17(12): 1188-92, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10585716

ABSTRACT

In utero injection of cationic liposome-DNA complexes (CLDCs) containing chloramphenicol acetyltransferase, beta-galactosidase (beta-gal), or human granulocyte colony-stimulating factor (hG-CSF) expression plasmids produced high-level gene expression in fetal rats. Tissues adjacent to the injection site exhibited the highest levels of gene expression. Chloramphenicol acetyltransferase expression persisted for at least 14 days and was reexpressed following postnatal reinjection of CLDCs. Intraperitoneal administration of the hG-CSF gene produced high serum hG-CSF levels. X-gal staining demonstrated widespread beta-gal expression in multiple fetal tissues and cell types. No toxic or inflammatory responses were observed, nor was there evidence of fetal-maternal or maternal-fetal gene transfer, suggesting that CLDCs may provide a useful alternative to viral vectors for in utero gene transfer.


Subject(s)
DNA/administration & dosage , Gene Transfer Techniques , Granulocyte Colony-Stimulating Factor/genetics , Animals , Blotting, Southern , Chloramphenicol O-Acetyltransferase/genetics , Female , Gene Expression , Germ Cells , Granulocyte Colony-Stimulating Factor/blood , Granulocyte Colony-Stimulating Factor/metabolism , Liposomes , Liver/metabolism , Plasmids , Polymerase Chain Reaction , Pregnancy , Rats , Rats, Inbred F344 , Uterus , beta-Galactosidase/genetics
11.
Hum Gene Ther ; 10(18): 2961-72, 1999 Dec 10.
Article in English | MEDLINE | ID: mdl-10609657

ABSTRACT

Local expression of cytokine genes by ex vivo transfection or intratumoral gene delivery can control the growth of cutaneous tumors. However, control of tumor metastases by conventional nonviral gene therapy approaches is more difficult. Intravenous injection of lipid-DNA complexes containing noncoding plasmid DNA can significantly inhibit the growth of early metastatic lung tumors. Therefore, we hypothesized that delivery of a cytokine gene by lipid-plasmid DNA complexes could induce even greater antitumor activity in mice with established lung metastases. The effectiveness of treatment with lipid-DNA complexes containing the IL-2 or IL-12 gene was compared with the effectiveness of treatment with complexes containing noncoding (empty vector) DNA. Treatment effects were evaluated in mice with either early (day 3) or late (day 6) established lung tumors. Lung tumor burdens and local intrapulmonary immune responses were assessed. Treatment with either noncoding plasmid DNA or with the IL-2 or IL-12 gene significantly inhibited the growth of early tumors. However, only treatment with the IL-2 or IL-12 gene induced a significant reduction in lung tumor burden in mice with more advanced metastases. Furthermore, the reduction in tumor burden was substantially greater than that achieved by treatment with recombinant cytokines. Treatment with the IL-2 or IL-12 gene was accompanied by increased numbers of NK cells and CD8+ T cells within lung tissues, increased cytotoxic activity, and increased local production of IFN-gamma by lung tissues, compared with treatment with noncoding DNA. Thus, cytokine gene delivery to the lungs by means of intravenously administered lipid-DNA complexes may be an effective method of controlling lung tumor metastases.


Subject(s)
Cell Division/genetics , Interleukin-12/genetics , Interleukin-2/genetics , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Animals , CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , DNA/administration & dosage , Genetic Vectors , Interferon-gamma/biosynthesis , Killer Cells, Natural/immunology , Lipids/administration & dosage , Lung/metabolism , Lung Neoplasms/immunology , Lymphocyte Depletion , Mice , Mice, Inbred Strains
12.
Hum Gene Ther ; 10(16): 2689-700, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10566897

ABSTRACT

We demonstrate here that intracerebroventricular or spinal cord (intrathecal) injection of either plasmid DNA alone or cationic liposome: DNA complexes (CLDCs) produces significant levels of expression of both reporter genes and biologically relevant genes in nonparenchymal cells lining both the brain and the spinal cord. Gene expression was identified both within the spinal cord and the brain after intracerebroventricular or intrathecal injection of either CLDCs or plasmid DNA alone. Intracerebroventricular or intrathecal injection of CLDCs containing the beta-galactosidase (beta-Gal) gene produced patchy, widely scattered areas of beta-Gal expression. The chloramphenicol acetyltransferase (CAT) reporter gene product reached peak levels between 24 hr and 1 week postinjection, and was still present at significant levels 3 weeks after a single intracerebroventricular or intrathecal injection. Intrathecal injection of the human granulocyte colony-stimulating factor (G-CSF) gene produced high levels of hG-CSF activity in both the spinal cord and the brain. Intracerebroventricular injection of CLDCs containing the murine nerve growth factor (NGF) gene increased mNGF levels in the hippocampus, a target region for cholinergic neurons in the medial septum, and increased cholinergic neurotransmitter synthetic enzyme choline acetyltransferase (ChAT) activity within the brain, a well-characterized effect of both purified and recombinant NGF protein. These findings indicate that intracerebroventricular or intrathecal injection of CLDCs can produce significant levels of expression of biologically and therapeutically relevant genes within the CNS. Efficient gene transfer into the CNS will facilitate the evaluation of gene function and regulation within the brain and spinal cord. We attempted to transfer and express genes within the brain and spinal cord by direct CNS injection of either DNA alone or CLDCs into the intraventricular and subarachnoid compartments. We show that intracerebroventricular or spinal cord (intrathecal) injection of either plasmid DNA alone or CLDCs produces significant levels of expression of both reporter genes and biologically relevant genes in nonparenchymal cells lining both the brain and the spinal cord. Intrathecal injection of the hG-CSF gene produced high levels of hG-CSF activity in both the spinal cord and the brain. Intracerebroventricular injection of CLDCs containing the murine NGF gene increased mNGF levels in the hippocampus, and increased cholinergic neurotransmitter synthetic enzyme ChAT activity within the brain. Locoregional diffusion of gene products expressed by transfected meningeal lining cells into brain and spinal cord parenchyma could potentially target secreted proteins within brain and spinal cord regions relevant to neuropathological states while limiting peripheral side effects.


Subject(s)
DNA/administration & dosage , DNA/analysis , Gene Expression Regulation , Spinal Cord/chemistry , Animals , Brain Chemistry , Central Nervous System/drug effects , Central Nervous System/metabolism , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , DNA/pharmacokinetics , Dosage Forms , Female , Gene Transfer Techniques , Genes, Reporter , Granulocyte Colony-Stimulating Factor/genetics , Granulocyte Colony-Stimulating Factor/metabolism , Humans , Injections, Intraventricular , Injections, Spinal , Liposomes , Mice , Mice, Inbred ICR , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , Plasmids , Tissue Distribution , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
13.
Hum Gene Ther ; 10(14): 2287-93, 1999 Sep 20.
Article in English | MEDLINE | ID: mdl-10515448

ABSTRACT

Perfluorochemical (PFC) liquids have both low surface tension and a high capacity to dissolve O2 and CO2, and have been shown to improve gas exchange and lung compliance in animal models of lung injury. We have previously demonstrated that perflubron and other PFC liquids enhance transgene expression in lungs of spontaneously breathing normal rodents after intratracheal instillation of either adenoviral or liposomal vectors followed by a single instillation of PFC liquid. We reasoned that PFC liquids may also be useful for enhancing transgene expression in abnormal lungs. GM-CSF knockout mice develop chronic accumulation of surfactant lipids and proteinaceous material in alveolar spaces and serve as a useful model of chronic alveolar filling. Intratracheal instillation of the adenoviral vector Adlac-Z resulted in patchy in situ distribution of beta-Gal activity, predominantly in larger proximal airways. In contrast, in mice instilled with Adlac-Z followed by instillation of a single dose of perflubron (10 ml/kg body weight), increased expression was observed in distal airway and alveolar epithelial cells. In particular, expression was observed in epithelial cells of debris-filled alveoli. Spectrophotometric measure of quantitative beta-Gal activity in lung homogenates demonstrated increased activity in lungs of mice receiving Adlac-Z plus perflubron compared with lungs of animals receiving Adlac-Z alone. These studies demonstrate that use of perflubron enhances transgene expression in lungs of animals with a chronic alveolar filling process. This approach may be applicable for gene delivery in diseases marked by chronic airway or alveolar filling such as cystic fibrosis.


Subject(s)
Fluorocarbons/pharmacology , Gene Expression/drug effects , Gene Transfer Techniques , Pulmonary Alveolar Proteinosis/therapy , Transgenes/drug effects , Adenoviridae/genetics , Animals , Fluorocarbons/chemistry , Genetic Vectors/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Hydrocarbons, Brominated , Liposomes , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Alveolar Proteinosis/enzymology , Pulmonary Alveolar Proteinosis/pathology , Pulmonary Alveoli/metabolism
14.
DNA Cell Biol ; 18(7): 533-47, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10433552

ABSTRACT

As the sequencing of the human genome proceeds, the need for a new screen for in vivo function is becoming apparent. Many investigators are turning to various transgenic models as a means of studying function. However, these approaches are very time consuming, with a transgene-expressing mouse model often taking months to establish. We have developed an efficient system for delivering genes in vivo, which allows the gene product to be studied as early as 24 h after introduction into the mouse model. The delivery system employs a novel cationic lipid, 1-[2-(9-(Z)-octadecenoyloxy)ethyl]-2-(8-(Z)-heptadecenyl)-3- (hydroxyethyl)imidazolinium chloride (DOTIM), and a neutral lipid, cholesterol, complexed with an expression vector containing the reporter gene chloramphenicol acetyl transferase (CAT). After a single intravenous injection of these complexes, several tissues were seen to express the transgene. High, persistent expression in the vascular endothelial cells in the mouse lung was obtained. Delivery of DNA in vivo has been evaluated by quantitative polymerase chain reaction and protein expression by CAT activity assays. In vivo studies showed reproducible expression in more than 500 mice injected via the tail vein. An early peak of expression was followed by lower, but sustained, expression for > 50 days. Transgene expression of CAT could also be identified by immunohistochemistry staining in mouse lung and appeared to be located within the capillaries. The pattern of in vivo expression could be modulated and targeted to specific organs by altering the lipid-DNA formulation. New expression vectors with altered introns and polyadenylation sites further improved expression. The expression reported here may be sufficient in magnitude, duration, and flexibility to be an attractive alternative, in some cases, to establishing transgenic animals by stable gene transfer.


Subject(s)
Gene Expression , Genetic Engineering/methods , Genetic Vectors , Imidazoles/metabolism , Lipid Metabolism , Transfection/methods , Animals , Animals, Genetically Modified , Chloramphenicol O-Acetyltransferase/analysis , Chloramphenicol O-Acetyltransferase/metabolism , Dose-Response Relationship, Drug , Female , Gene Targeting , Genes, Reporter , Granulocyte Colony-Stimulating Factor/metabolism , Lipids , Lung/anatomy & histology , Lung/metabolism , Mice , Pancreas/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spleen/metabolism , Time Factors
15.
Histochem J ; 31(4): 231-6, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10447064

ABSTRACT

Minces of several organs from the transgenic mouse ROSAbeta-gal 26 (ROSA-26), which robustly expresses bacterial lac-Z in most tissues, were exposed to 4-bromo-5-chloro-3-indoyl-beta-D-galactopyrosanide (X-gal) at pH ranging from 7.5 to 9.5 to determine the optimal pH for in situ demonstration of bacterial beta-galactosidase activity (neutral pH optimum) while minimizing detection of potentially confounding endogenous mammalian beta-galactosidase (acidic pH optimum). Similar studies were performed with organ minces from C57BL/6 mice, Sprague-Dawley rats, New Zealand white rabbits, and macaques to confirm the effect of pH on minimizing detection of endogenous mammalian beta-galactosidase. In all organs evaluated; heart, liver, spleen, kidney, brain, and skeletal muscle, endogenous beta-galactosidase activity was rarely detected following incubation at pH greater than 7.5. In contrast, bacterial beta-galactosidase activity in the ROSA-26 mice was strongly detected in organ minces following incubation at pH 8.0-9.0. These findings are similar to previous observations we have made in lung minces and confirm that a simple alteration of a commonly used histochemical technique for detecting in situ beta-galactosidase activity, raising the reaction buffer pH to weakly alkaline range, can reliably distinguish between endogenous activity and that resulting from exogenous bacterial gene expression.


Subject(s)
beta-Galactosidase/analysis , Animals , Brain/enzymology , Galactosides/metabolism , Gene Expression , Histocytochemistry/methods , Hydrogen-Ion Concentration , Indoles/metabolism , Kidney/enzymology , Lac Operon , Macaca , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Skeletal/enzymology , Myocardium/enzymology , Rabbits , Rats , Rats, Sprague-Dawley , Spleen/enzymology
16.
J Immunol ; 163(3): 1552-61, 1999 Aug 01.
Article in English | MEDLINE | ID: mdl-10415059

ABSTRACT

Cationic lipid-DNA complexes (CLDC) are reported to be safe and effective for systemic gene delivery, particularly to the lungs. However, we observed that i.v. injection of CLDC induced immunologic effects not previously reported. We found that even very low doses of CLDC administered i.v. induced marked systemic immune activation. This response included strong up-regulation of CD69 expression on multiple cell types and systemic release of high levels of Th1 cytokines, from both lung and spleen mononuclear cells. CLDC were much more potent immune activators on a per weight basis than either LPS or poly(I:C). The remarkable potency of CLDC appeared to result from enhancement of the immune stimulatory properties of DNA, since cationic lipids alone were without immune stimulatory activity. Systemic treatment with CLDC controlled tumor growth and significantly prolonged survival times in mice with metastatic pulmonary tumors. NK cells accumulated to high levels in the lungs of CLDC-treated mice, were functionally activated, and released high levels of IFN-gamma. The antitumor activity induced by CLDC injection was dependent on both NK cells and IFN-gamma. Thus, DNA complexed to cationic liposomes becomes highly immunostimulatory and capable of inducing strong antitumor activity when administered systemically.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antineoplastic Agents/immunology , DNA, Bacterial/immunology , Liposomes/immunology , Lymphocyte Activation/immunology , Animals , Antineoplastic Agents/administration & dosage , Cell Division/immunology , Cytotoxicity, Immunologic/immunology , DNA, Bacterial/administration & dosage , Dose-Response Relationship, Immunologic , Drug Combinations , Female , Immunity, Innate , Injections, Intravenous , Interferon-gamma/immunology , Interferon-gamma/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Liposomes/administration & dosage , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Tumor Cells, Cultured
17.
Exp Lung Res ; 25(4): 317-33, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10378103

ABSTRACT

Perfluorochemical (PFC) liquids have been shown to improve gas exchange and lung compliance in models of lung injury. We reasoned they may also be useful as a vehicle for gene transfer by improving transgene distribution throughout the lung as well as increasing total transgene expression. We have developed a model for PFC liquid use in spontaneously breathing rodents that obviates the need for intubation and ventilation. Intratracheal instillation of the adenoviral vector Adlac-Z resulted in patchy distribution of beta-galactosidase (beta-gal) activity as demonstrated using X-gal histochemistry. In contrast, in rats instilled with Adlac-Z followed by instillation of PFC liquid, more uniformly distributed and increased beta-gal activity was observed. Activity in distal airway and alveolar epithelium was particularly increased. Quantitative measure of beta-gal activity in lung homogenates demonstrated a 3- to 6-fold increase in total activity in lungs of rats receiving Adlac-Z and PFC liquid compared to animals receiving Adlac-Z alone. These studies show that PFC liquids can enhance both the distribution and the total amount of transgene expressed following adenoviral-mediated vector transfer to lungs during spontaneous breathing. Use of PFC liquids may increase the efficacy of gene transfer strategies for treatment of cystic fibrosis and other lung diseases.


Subject(s)
Adenoviridae/genetics , Fluorocarbons/pharmacology , Furans/pharmacology , Gene Expression/drug effects , Genetic Vectors , Lung/enzymology , Respiration , beta-Galactosidase/genetics , Animals , Defective Viruses , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Fluorocarbons/administration & dosage , Furans/administration & dosage , Gene Transfer Techniques , Lac Operon/genetics , Lung/diagnostic imaging , Lung/physiology , Male , Mice , Mice, Inbred C57BL , Radiography , Rats , Rats, Sprague-Dawley , beta-Galactosidase/metabolism
18.
J Invest Dermatol ; 112(3): 370-5, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10084316

ABSTRACT

We topically applied naked plasmid DNA containing the luciferase or chloramphenicol acetyltransferase cDNA directly to mouse skin. Gene expression was detected in skin samples as early as 4 h after DNA application, plateaued from 16 to 72 h post-application, and had decreased significantly by 7 d post-application. Reporter gene activity following topical DNA delivery was comparable with that produced by intradermal injection of DNA. Plasmid DNA at concentrations > or =0.25 microg per microl were required to achieve maximal expression levels. Reporter gene expression following topical administration was largely confined to the superficial layers of the epidermis and to hair follicles. Surprisingly, certain cationic liposomes inhibited the efficiency of cutaneous gene transfer. This technique provides a simple, clinically relevant approach to deliver genes to the skin, with potential application in treating a variety of cutaneous disorders.


Subject(s)
Gene Transfer Techniques , Skin Physiological Phenomena , Administration, Topical , Animals , Cations/pharmacology , Chloramphenicol O-Acetyltransferase/genetics , DNA/administration & dosage , Epidermis/physiology , Female , Gene Expression/drug effects , Gene Expression/physiology , Hair Follicle/physiology , Liposomes/pharmacology , Luciferases/genetics , Mice , Mice, Inbred ICR , Pharmaceutical Vehicles , Plasmids/genetics , Time Factors
19.
Hum Gene Ther ; 8(13): 1545-54, 1997 Sep 01.
Article in English | MEDLINE | ID: mdl-9322087

ABSTRACT

Bacterial lacZ is one of the most commonly used reporter genes for assessing gene transfer to lung. However, lung contains endogenous beta-galactosidase (beta-Gal), which can confound estimation of exogenous lacZ expression by histochemical techniques (i.e., X-Gal) for in situ demonstration of enzyme activity. We investigated several parameters of the X-Gal reaction, including time and temperature of X-Gal exposure as well as lung tissue processing and fixation techniques, and found that none of these could be used to distinguish between endogenous and exogenous beta-Gal activities. The mammalian and bacterial beta-Gal enzymes, however, have pH optima in the acidic and neutral ranges, respectively. Exposing whole lung, lung minces, or mounted frozen sections of lung to X-Gal at mildly alkaline pH (pH 8.0-8.5), minimized detection of endogenous activity in lungs from a variety of species while preserving that resulting from bacterial enzyme activity in a transgenic mouse expressing lacZ. This technique was also useful in distinguishing endogenous activity from that resulting from adenovirus-mediated lacZ gene transfer to diploid lung fibroblasts in primary culture. An appropriate buffer that maintains the desired pH throughout the duration of X-Gal exposure must be used.


Subject(s)
Gene Expression , Lac Operon , Lung/enzymology , beta-Galactosidase/metabolism , Animals , Bacteria/enzymology , Genes, Reporter , Histocytochemistry , Humans , Hydrogen-Ion Concentration , Mice , Mice, Inbred Strains , Papio , Rats , Rats, Sprague-Dawley , Species Specificity , Tissue Fixation , Tumor Cells, Cultured , beta-Galactosidase/genetics
20.
Clin Cancer Res ; 3(3): 339-45, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9815690

ABSTRACT

Clinical trials of immunotoxins in cancer patients have been limited in many cases by vascular leak syndrome (VLS). Recently, rats were identified as a model for VLS induced by BR96 sFv-PE40, a carcinoma-reactive single-chain immunotoxin. In this study, the toxin component of this immunotoxin, PE40, was found to be responsible for inducing hydrothorax in rats, thereby demonstrating that direct binding to the BR96 antigen was not essential to the onset of VLS. Mutational analysis of PE40 determined that both ADP ribosylation and proteolytic processing functions innate to Pseudomonas exotoxin A (PE) were necessary for PE40 to induce hydrothorax in rats; however, neither function by itself was sufficient for VLS induction. Additionally, nonsteroidal anti-inflammatory agents were found to block VLS in rats receiving BR96 sFv-PE40. These results demonstrate that the toxin component of PE-based immunotoxins induce VLS and suggest agents for clinical management of the toxicity.


Subject(s)
ADP Ribose Transferases , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antineoplastic Agents/toxicity , Bacterial Toxins , Capillary Leak Syndrome/chemically induced , Capillary Leak Syndrome/prevention & control , Dexamethasone/therapeutic use , Exotoxins/toxicity , Gene Expression Regulation/drug effects , Hydrothorax/chemically induced , Hydrothorax/prevention & control , Immunotoxins/toxicity , Lung/drug effects , Transcription, Genetic/drug effects , Virulence Factors , Animals , Antibodies, Monoclonal , Cyclooxygenase 1 , Cyclooxygenase 2 , Female , Interleukin-1/genetics , Isoenzymes/genetics , Lung/metabolism , Lung/pathology , Membrane Proteins , Prostaglandin-Endoperoxide Synthases/genetics , Rats , Rats, Sprague-Dawley , Rats, Wistar , Recombinant Fusion Proteins/toxicity , Pseudomonas aeruginosa Exotoxin A
SELECTION OF CITATIONS
SEARCH DETAIL
...