Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
2.
Mol Ther Methods Clin Dev ; 28: 146-159, 2023 Mar 09.
Article in English | MEDLINE | ID: mdl-36654797

ABSTRACT

The adeno-associated viral vector (AAV) provides a safe and efficient gene therapy platform with several approved products that have marked therapeutic impact for patients. However, a major bottleneck in the development and commercialization of AAV remains the efficiency, cost, and scalability of AAV production. Chromatographic methods have the potential to allow purification at increased scales and lower cost but often require optimization specific to each serotype. Here, we demonstrate that the POROS CaptureSelect AAVX affinity resin efficiently captures a panel of 15 divergent AAV serotypes, including the commonly used AAV2, AAV8, AAV9, PHP.B, and Anc80. We also find that AAVX resin can be regenerated repeatedly without loss of efficiency or carry-over contamination. While AAV preps purified with AAVX showed a higher fraction of empty capsids than preps purified using iodixanol ultracentrifugation, the potency of the AAVX purified vectors was comparable with that of iodixanol purified vectors both in vitro and in vivo. Finally, optimization of the purification protocol resulted in a process with an overall efficiency of 65%-80% across all scales and AAV serotypes tested. These data establish AAVX affinity chromatography as a versatile and efficient method for purification of a broad range of AAV serotypes.

3.
Nat Commun ; 13(1): 1359, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35292639

ABSTRACT

Inner ear gene therapy using adeno-associated viral vectors (AAV) promises to alleviate hearing and balance disorders. We previously established the benefits of Anc80L65 in targeting inner and outer hair cells in newborn mice. To accelerate translation to humans, we now report the feasibility and efficiency of the surgical approach and vector delivery in a nonhuman primate model. Five rhesus macaques were injected with AAV1 or Anc80L65 expressing eGFP using a transmastoid posterior tympanotomy approach to access the round window membrane after making a small fenestra in the oval window. The procedure was well tolerated. All but one animal showed cochlear eGFP expression 7-14 days following injection. Anc80L65 in 2 animals transduced up to 90% of apical inner hair cells; AAV1 was markedly less efficient at equal dose. Transduction for both vectors declined from apex to base. These data motivate future translational studies to evaluate gene therapy for human hearing disorders.


Subject(s)
Dependovirus , Genetic Vectors , Animals , Cochlea/physiology , Dependovirus/genetics , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors/genetics , Macaca mulatta/genetics , Mice
4.
Mol Ther Methods Clin Dev ; 17: 107-121, 2020 Jun 12.
Article in English | MEDLINE | ID: mdl-31909084

ABSTRACT

Generation and screening of libraries of adeno-associated virus (AAV) variants have emerged as a powerful method for identifying novel capsids for gene therapy applications. For the majority of libraries, vast population diversity requires multiplexed production, in which a library of inverted terminal repeat (ITR)-containing plasmid variants is transfected together into cells to generate the viral library. This process has the potential to be confounded by cross-packaging and mosaicism, in which particles are comprised of genomes and capsid monomers derived from different library members. Here, we investigate the prevalence of cross-packaging and mosaicism in simplified, minimal libraries using novel assays designed to assess capsid composition and packaging fidelity. We show that AAV library variants are prone to cross-packaging and capsid mosaic formation when produced at high plasmid levels, although to a lesser extent than in a recombinant context. We also provide experimental evidence that dilution of input library DNA significantly increases capsid monomer homogeneity and increases capsid:genome correlation in AAV libraries. Lastly, we determine that similar dilution methods yield higher-quality libraries when used for in vivo screens. Together, these findings quantitatively characterized the prevalence of cross-packaging and mosaicism in AAV libraries and established conditions that minimize related noise in subsequent screens.

5.
Sci Rep ; 7: 46581, 2017 04 21.
Article in English | MEDLINE | ID: mdl-28429726

ABSTRACT

Verteporfin (VP) was first used in Photodynamic therapy, where a non-thermal laser light (689 nm) in the presence of oxygen activates the drug to produce highly reactive oxygen radicals, resulting in local cell and tissue damage. However, it has also been shown that Verteporfin can have non-photoactivated effects such as interference with the YAP-TEAD complex of the HIPPO pathway, resulting in growth inhibition of several neoplasias. More recently, it was proposed that, another non-light mediated effect of VP is the formation of cross-linked oligomers and high molecular weight protein complexes (HMWC) that are hypothesized to interfere with autophagy and cell growth. Here, in a series of experiments, using human uveal melanoma cells (MEL 270), human embryonic kidney cells (HEK) and breast cancer cells (MCF7) we showed that Verteporfin-induced HMWC require the presence of light. Furthermore, we showed that the mechanism of this cross-linking, which involves both singlet oxygen and radical generation, can occur very efficiently even after lysis of the cells, if the lysate is not protected from ambient light. This work offers a better understanding regarding VP's mechanisms of action and suggests caution when one studies the non-light mediated actions of this drug.


Subject(s)
Light , Multiprotein Complexes/metabolism , Neoplasms , Photochemotherapy , Photosensitizing Agents , Verteporfin , Adaptor Proteins, Signal Transducing/metabolism , DNA-Binding Proteins/metabolism , HEK293 Cells , Hippo Signaling Pathway , Humans , MCF-7 Cells , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Phosphoproteins/metabolism , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/pharmacology , Protein Serine-Threonine Kinases/metabolism , Transcription Factors , Verteporfin/pharmacokinetics , Verteporfin/pharmacology , YAP-Signaling Proteins
7.
BMC Biol ; 14: 59, 2016 07 13.
Article in English | MEDLINE | ID: mdl-27412481

ABSTRACT

BACKGROUND: Type IV collagen is the main component of the basement membrane that gives strength to the blood-gas barrier (BGB). In mammals, the formation of a mature BGB occurs primarily after birth during alveologenesis and requires the formation of septa from the walls of the saccule. In contrast, in avians, the formation of the BGB occurs rapidly and prior to hatching. Mutation in basement membrane components results in an abnormal alveolar phenotype; however, the specific role of type IV collagen in regulating alveologenesis remains unknown. RESULTS: We have performed a microarray expression analysis in late chick lung development and found that COL4A1 and COL4A2 were among the most significantly upregulated genes during the formation of the avian BGB. Using mouse models, we discovered that mutations in murine Col4a1 and Col4a2 genes affected the balance between lung epithelial progenitors and differentiated cells. Mutations in Col4a1 derived from the vascular component were sufficient to cause defects in vascular development and the BGB. We also show that Col4a1 and Col4a2 mutants displayed disrupted myofibroblast proliferation, differentiation and migration. Lastly, we revealed that addition of type IV collagen protein induced myofibroblast proliferation and migration in monolayer culture and increased the formation of mesenchymal-epithelial septal-like structures in co-culture. CONCLUSIONS: Our study showed that type IV collagen and, therefore the basement membrane, play fundamental roles in coordinating alveolar morphogenesis. In addition to its role in the formation of epithelium and vasculature, type IV collagen appears to be key for alveolar myofibroblast development by inducing their proliferation, differentiation and migration throughout the developing septum.


Subject(s)
Collagen Type IV/metabolism , Endothelial Cells/cytology , Epithelial Cells/cytology , Morphogenesis , Peptide Fragments/metabolism , A549 Cells , Animals , Basement Membrane/metabolism , Cell Differentiation , Cell Movement , Cell Proliferation , Cells, Cultured , Chick Embryo , Coculture Techniques , Collagen Type IV/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Lung/cytology , Mice , Mice, Knockout , Microarray Analysis , Mutation , Myofibroblasts/cytology , Peptide Fragments/genetics , Up-Regulation
8.
Invest Ophthalmol Vis Sci ; 55(7): 4175-85, 2014 Apr 29.
Article in English | MEDLINE | ID: mdl-24781943

ABSTRACT

PURPOSE: To evaluate the effects and mechanism of aminoimidazole carboxamide ribonucleotide (AICAR), an AMP-dependent kinase (AMPK) activator, on the growth of uveal melanoma cell lines. METHODS: Four different cell lines were treated with AICAR (1-4 mM). Cell growth was assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium (MTT) assay. Cell cycle analysis was conducted by flow cytometry; additionally, expression of cell-cycle control proteins, cell growth transcription factors, and downstream effectors of AMPK were determined by RT-PCR and Western blot. RESULTS: Aminoimidazole carboxamide ribonucleotide inhibited cell growth, induced S-phase arrest, and led to AMPK activation. Aminoimidazole carboxamide ribonucleotide treatment was associated with inhibition of eukaryotic translation initiation factor 4E-BP1 phosphorylation, a marker of mammalian target of rapamycin (mTOR) pathway activity. Aminoimidazole carboxamide ribonucleotide treatment was also associated with downregulation of cyclins A and D, but had minimal effects on the phosphorylation of ribosomal protein S6 or levels of the macroautophagy marker LC3B. The effects of AICAR were abolished by treatment with dipyridamole, an adenosine transporter inhibitor that blocks the entry of AICAR into cells. Treatment with adenosine kinase inhibitor 5-iodotubericidin, which inhibits the conversion of AICAR to its 5'-phosphorylated ribotide 5-aminoimidazole-4-carboxamide-1-D-ribofuranosyl-5'-monophosphate (ZMP; the direct activator of AMPK), reversed most of the growth-inhibitory effects, indicating that some of AICAR's antiproliferative effects are mediated at least partially through AMPK activation. CONCLUSIONS: Aminoimidazole carboxamide ribonucleotide inhibited uveal melanoma cell proliferation partially through activation of the AMPK pathway and downregulation of cyclins A1 and D1.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic , Melanoma/genetics , Phosphotransferases (Phosphate Group Acceptor)/metabolism , RNA, Neoplasm/genetics , Ribonucleotides/pharmacology , TOR Serine-Threonine Kinases/genetics , Uveal Neoplasms/genetics , Aminoimidazole Carboxamide/pharmacology , Blotting, Western , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Flow Cytometry , Humans , Melanoma/drug therapy , Melanoma/enzymology , Phosphotransferases (Phosphate Group Acceptor)/drug effects , Real-Time Polymerase Chain Reaction , TOR Serine-Threonine Kinases/metabolism , Uveal Neoplasms/drug therapy , Uveal Neoplasms/enzymology
9.
Gynecol Oncol ; 120(1): 128-34, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21056908

ABSTRACT

OBJECTIVE: Given the fact that Mullerian Inhibiting Substance (MIS) causes complex remodeling of the urogenital ridge and regression of the Mullerian ducts during male embryonic development, we examined whether MIS could affect similar cell properties such as migration and invasion that could contribute ultimately to micro-metastasis of cancers arising from Mullerian tissues. MIS receptor expressing cell lines found to be invasive and migratory in vivo are examined in an in vivo assay that is cost-effective. METHODS: We designed in vitro and in vivo experiments to determine if MIS inhibited the movement of cancer lines IGROV-1, HEp3, MDA-MB-231, and HT1080 in cell culture invasion/migration chamber assays and in chick embryo metastasis assays. RESULTS: MIS, at concentrations below those that inhibit cell proliferation, blocked in vitro invasion and in vivo migration of epithelial cancer cells that express the MIS receptor. CONCLUSIONS: While our laboratory has previously established MIS as an inhibitor of cancer cell proliferation using in vitro assays and in vivo xenografts, we now show that MIS can also inhibit in vivo tumor migration.


Subject(s)
Anti-Mullerian Hormone/pharmacology , Cell Movement/drug effects , Neoplasms/drug therapy , Animals , Anti-Mullerian Hormone/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Chick Embryo , Chorioallantoic Membrane/cytology , Chorioallantoic Membrane/drug effects , Dose-Response Relationship, Drug , Female , Fibrosarcoma/drug therapy , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Neoplasm Invasiveness , Neoplasms/metabolism , Neoplasms/pathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Paclitaxel/administration & dosage , Receptors, Peptide/metabolism , Receptors, Transforming Growth Factor beta/metabolism
10.
FASEB J ; 24(8): 2620-30, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20371623

ABSTRACT

5-Aminoimidazole-4-carboxamide-1-beta-4-ribofuranoside (AICAR), an analog of AMP, is widely used as an activator of AMP-kinase (AMPK), a protein that regulates the responses of the cell to energy change. We studied the effects of AICAR on the growth of retinoblastoma cell lines (Y79, WERI, and RB143). AICAR inhibited Rb cell growth, induced apoptosis and S-phase cell cycle arrest, and led to activation of AMPK. These effects were abolished by treatment with dypiridamole, an inhibitor that blocks entrance of AICAR into cells. Treatment with the adenosine kinase inhibitor 5-iodotubericidin to inhibit the conversion of AICAR to ZMP (the direct activator of AMPK) reversed most of the growth-inhibiting effects of AICAR, indicating that some of the antiproliferative effects of AICAR are mediated through AMPK activation. In addition, AICAR treatment was associated with inhibition of the mammalian target of rapamycin pathway, decreased phosphorylation of ribosomal protein-S6 and 4E-BP1, down-regulation of cyclins A and E, and decreased expression of p21. Our results indicate that AICAR-induced activation of AMPK inhibits retinoblastoma cell growth. This is one of the first descriptions of a nonchemotherapeutic drug with low toxicity that may be effective in treating Rb patients.


Subject(s)
Adenylate Kinase/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Retinoblastoma/drug therapy , Ribonucleotides/pharmacology , Aminoimidazole Carboxamide/pharmacology , Antineoplastic Agents , Apoptosis , Cell Cycle , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Hypoglycemic Agents , Phosphorylation/drug effects , Retinoblastoma/pathology , Signal Transduction/drug effects
11.
Protein Expr Purif ; 70(1): 32-8, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19755162

ABSTRACT

Mullerian Inhibiting Substance (MIS), a member of the TGF-beta family, causes regression of the Mullerian duct in male embryos, after binding to Mullerian Inhibiting Substance Receptor II (MISRII). It has also been extensively demonstrated that it can inhibit proliferation of various cancer cell lines such as ovarian, prostate, and breast cancer in vitro and in vivo. Hence, the availability of a recombinant, epitope tagged, bioactive MIS is important for the selection of patients for treatment and for probing novel molecular targets for MIS in various tissues. To this end, we have expressed a recombinant, internally FLAG-tagged form of hMIS with the tag (DYKDDDDK) immediately after the cleavage site (427-428) of MIS at the C-terminus with a modified dibasic cleavage motif sequence. We show that this construct results in a highly pure, endogenously processed (cleaved) FLAG MIS, that causes complete regression of the Mullerian Duct in an organ culture assay. In addition, purified FLAG MIS was able to bind and affinity purify both transfected and endogenous MIS type II receptor. The availability of this fully functional, epitope tagged form of MIS should facilitate scale-up for preclinical and clinical use and should also be used for the study of MIS binding proteins and for tracking in pharmacokinetic studies.


Subject(s)
Anti-Mullerian Hormone/genetics , Recombinant Fusion Proteins/chemistry , Amino Acid Sequence , Anti-Mullerian Hormone/chemistry , Anti-Mullerian Hormone/metabolism , Blotting, Western , Humans , Molecular Sequence Data , Oligopeptides , Peptides/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism
12.
Blood ; 101(10): 4033-41, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12576324

ABSTRACT

Hairy cell leukemia (HCL) is a chronic lymphoproliferative disease, the cause of which is unknown. Diagnostic of HCL is abnormal expression of the gene that encodes the beta2 integrin CD11c. In order to determine the cause of CD11c gene expression in HCL the CD11c gene promoter was characterized. Transfection of the CD11c promoter linked to a luciferase reporter gene indicated that it is sufficient to direct expression in hairy cells. Mutation analysis demonstrated that of predominant importance to the activity of the CD11c promoter is its interaction with the activator protein-1 (AP-1) family of transcription factors. Comparison of nuclear extracts prepared from hairy cells with those prepared from other cell types indicated that hairy cells exhibit abnormal constitutive expression of an AP-1 complex containing JunD. Functional inhibition of AP-1 expressed by hairy cells reduced CD11c promoter activity by 80%. Inhibition of Ras, which represents an upstream activator of AP-1, also significantly inhibited the CD11c promoter. Furthermore, in the hairy cell line EH, inhibition of Ras signaling through mitogen-activated protein kinase/extracellular signal-regulated kinase kinases 1 and 2 (MEK1/2) reduced not only CD11c promoter activity but also reduced both CD11c surface expression and proliferation. Expression in nonhairy cells of a dominant-positive Ras mutant activated the CD11c promoter to levels equivalent to those in hairy cells. Together, these data indicate that the abnormal expression of the CD11c gene characteristic of HCL is dependent upon activation of the proto-oncogenes ras and junD.


Subject(s)
CD11c Antigen/genetics , Gene Expression Regulation, Neoplastic , Genes, jun , Genes, ras , Leukemia, Hairy Cell/genetics , Promoter Regions, Genetic , Proto-Oncogenes , Antigens, CD/genetics , Base Sequence , Binding Sites , Butadienes/pharmacology , Cell Division/drug effects , Enzyme Inhibitors/pharmacology , HeLa Cells , Humans , Leukemia, Hairy Cell/immunology , Leukemia, Hairy Cell/pathology , Molecular Sequence Data , Nitriles/pharmacology , Plasmids , Transcription Factor AP-1/metabolism , Tumor Cells, Cultured , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...