22 results on '"Nicholas W. Keiser"'
Search Results
2. Chondroitin Sulfate is the Primary Receptor for a Peptide-Modified AAV That Targets Brain Vascular Endothelium In Vivo
- Author
-
James C Geoghegan, Nicholas W Keiser, Anna Okulist, Inês Martins, Matthew S Wilson, and Beverly L Davidson
- Subjects
AAV ,brain vasculature ,capsid engineering ,chondroitin sulfate ,endothelium ,phage display ,receptor ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Recently, we described a peptide-modified AAV2 vector (AAV-GMN) containing a capsid-displayed peptide that directs in vivo brain vascular targeting and transduction when delivered intravenously. In this study, we sought to identify the receptor that mediates transduction by AAV-GMN. We found that AAV-GMN, but not AAV2, readily transduces the murine brain endothelial cell line bEnd.3, a result that mirrors previously observed in vivo transduction profiles of brain vasculature. Studies in vitro revealed that the glycosaminoglycan, chondroitin sulfate C, acts as the primary receptor for AAV-GMN. Unlike AAV2, chondroitin sulfate expression is required for cell transduction by AAV-GMN, and soluble chondroitin sulfate C can robustly inhibit AAV-GMN transduction of brain endothelial cells. Interestingly, AAV-GMN retains heparin-binding properties, though in contrast to AAV2, it poorly transduces cells that express heparan sulfate but not chondroitin sulfate, indicating that the peptide insertion negatively impacts heparan-mediated transduction. Lastly, when delivered directly, this modified virus can transduce multiple brain regions, indicating that the potential of AAV-GMN as a therapeutic gene delivery vector for central nervous system disorders is not restricted to brain vascular endothelium.
- Published
- 2014
- Full Text
- View/download PDF
3. Infection Is Not Required for Mucoinflammatory Lung Disease in CFTR-Knockout Ferrets
- Author
-
Chandler W. Jensen-Cody, Yalan Li, Xingshen Sun, T. Idil Apak Evans, J. Kirk Harris, Scott R. Tyler, Nicholas W. Keiser, Weihong Zhou, Yulong Zhang, John F. Engelhardt, Anthony M. Swatek, Kai Wang, Bradley H. Rosen, Pavana G. Rotti, Nan He, Preston J. Anderson, Bo Liang, R. Marshall Pope, Katherine N. Gibson-Corley, Eric A. Hoffman, Leonard A Brooks, Shashanna R. Moll, Jaimie S. Gray, Kalpaj R. Parekh, and Maheen Rajput
- Subjects
Lung Diseases ,0301 basic medicine ,Pulmonary and Respiratory Medicine ,Inflammatory response ,Cystic Fibrosis Transmembrane Conductance Regulator ,Inflammation ,Infections ,Critical Care and Intensive Care Medicine ,Cystic fibrosis ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Lung ,Respiratory Tract Infections ,business.industry ,Disease progression ,Ferrets ,Airway obstruction ,medicine.disease ,Mucus ,Disease Models, Animal ,030104 developmental biology ,030228 respiratory system ,Lung disease ,Immunology ,medicine.symptom ,business - Abstract
Classical interpretation of cystic fibrosis (CF) lung disease pathogenesis suggests that infection initiates disease progression, leading to an exuberant inflammatory response, excessive mucus, and ultimately bronchiectasis. Although symptomatic antibiotic treatment controls lung infections early in disease, lifelong bacterial residence typically ensues. Processes that control the establishment of persistent bacteria in the CF lung, and the contribution of noninfectious components to disease pathogenesis, are poorly understood.To evaluate whether continuous antibiotic therapy protects the CF lung from disease using a ferret model that rapidly acquires lethal bacterial lung infections in the absence of antibiotics.CFTR (cystic fibrosis transmembrane conductance regulator)-knockout ferrets were treated with three antibiotics from birth to several years of age and lung disease was followed by quantitative computed tomography, BAL, and histopathology. Lung disease was compared with CFTR-knockout ferrets treated symptomatically with antibiotics.Bronchiectasis was quantified from computed tomography images. BAL was evaluated for cellular differential and features of inflammatory cellular activation, bacteria, fungi, and quantitative proteomics. Semiquantitative histopathology was compared across experimental groups. We demonstrate that lifelong antibiotics can protect the CF ferret lung from infections for several years. Surprisingly, CF animals still developed hallmarks of structural bronchiectasis, neutrophil-mediated inflammation, and mucus accumulation, despite the lack of infection. Quantitative proteomics of BAL from CF and non-CF pairs demonstrated a mucoinflammatory signature in the CF lung dominated by Muc5B and neutrophil chemoattractants and products.These findings implicate mucoinflammatory processes in the CF lung as pathogenic in the absence of clinically apparent bacterial and fungal infections.
- Published
- 2018
- Full Text
- View/download PDF
4. Glandular Proteome Identifies Antiprotease Cystatin C as a Critical Modulator of Airway Hydration and Clearance
- Author
-
Scott R. Tyler, Nam Soo Joo, Hyung Ju Cho, Jordy J. Hsiao, Michael E. Wright, T. Idil Apak Evans, Weiliang Xie, Yulong Zhang, Ziying Yan, Nicholas W. Keiser, John F. Engelhardt, and Jeffrey J. Wine
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Epithelial sodium channel ,medicine.medical_specialty ,Cystic Fibrosis ,Proteome ,Mucociliary clearance ,Clinical Biochemistry ,Bronchi ,Cystic fibrosis ,03 medical and health sciences ,Internal medicine ,medicine ,Animals ,Humans ,Cystatin C ,Molecular Biology ,Original Research ,Cathepsin S ,Submucosal glands ,biology ,Chemistry ,Ferrets ,Cell Biology ,respiratory system ,medicine.disease ,Cystic fibrosis transmembrane conductance regulator ,respiratory tract diseases ,Cell biology ,Trachea ,HEK293 Cells ,030104 developmental biology ,Endocrinology ,Chloride channel ,biology.protein - Abstract
Defects in the cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel lead to viscous secretions from submucosal glands that cannot be properly hydrated and cleared by beating cilia in cystic fibrosis (CF) airways. The mechanisms by which CFTR, and the predominant epithelial sodium channel (ENaC), control the hydration and clearance of glandular secretions remain unclear. We used a proteomics approach to characterize the proteins contained in CF and non-CF submucosal gland fluid droplets and found that differentially regulated proteases (cathepsin S and H) and their antiprotease (cystatin C) influenced the equilibration of fluid on the airway surface and tracheal mucociliary clearance (MCC). Contrary to prevailing models of airway hydration and clearance, cystatin C, or raising the airway surface liquid (ASL) pH, inhibited cathepsin-dependent ENaC-mediated fluid absorption and raised the height of ASL, and yet decreased MCC velocity. Importantly, coupling of both CFTR and ENaC activities were required for effective MCC and for effective ASL height equilibration after volume challenge. Cystatin C–inhibitable cathepsins controlled initial phases of ENaC-mediated fluid absorption, whereas CFTR activity was required to prevent ASL dehydration. Interestingly, CF airway epithelia absorbed fluid more slowly owing to reduced cysteine protease activity in the ASL but became abnormally dehydrated with time. Our findings demonstrate that, after volume challenge, pH-dependent protease-mediated coupling of CFTR and ENaC activities are required for rapid fluid equilibration at the airway surface and for effective MCC. These findings provide new insights into how glandular fluid secretions may be equilibrated at the airway surface and how this process may be impaired in CF.
- Published
- 2016
- Full Text
- View/download PDF
5. Gastrointestinal Pathology in Juvenile and Adult CFTR-Knockout Ferrets
- Author
-
Nicholas W. Keiser, Joann M. Kinyon, Thomas J. Lynch, Weihong Zhou, John F. Engelhardt, Xingshen Sun, Yaling Yi, Scott R. Tyler, J. Adam Goeken, Yulong Zhang, Yi Song, Timothy S. Frana, Xiaoming Liu, Lucas R. Hoffman, Kalpaj R. Parekh, Mitchell J. Brittnacher, Weiliang Xie, Alicia K. Olivier, Hongshu Sui, Bo Liang, David K. Meyerholz, Xiaoyan Wang, Samuel I. Miller, Danielle Fligg, Kyle R. Hager, John T. Fisher, Zoe A. Stewart, Paul M. Kaminsky, Ziying Yan, Christopher E. Pope, Hillary S. Hayden, and Matthew C. Radey
- Subjects
Aging ,medicine.medical_specialty ,Pathology ,Cystic Fibrosis ,Cystic Fibrosis Transmembrane Conductance Regulator ,Gastroenterology ,Cystic fibrosis ,Pathology and Forensic Medicine ,Gene Knockout Techniques ,Atrophy ,Fibrosis ,Internal medicine ,medicine ,Animals ,Humans ,Gastrointestinal tract ,Bacteria ,biology ,Biliary hyperplasia ,Gallbladder ,Ferrets ,Regular Article ,Gastrointestinal pathology ,medicine.disease ,Cystic fibrosis transmembrane conductance regulator ,3. Good health ,Gastrointestinal Tract ,Mucus ,medicine.anatomical_structure ,Organ Specificity ,biology.protein - Abstract
Cystic fibrosis (CF) is a multiorgan disease caused by loss of a functional cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel in many epithelia of the body. Here we report the pathology observed in the gastrointestinal organs of juvenile to adult CFTR-knockout ferrets. CF gastrointestinal manifestations included gastric ulceration, intestinal bacterial overgrowth with villous atrophy, and rectal prolapse. Metagenomic phylogenetic analysis of fecal microbiota by deep sequencing revealed considerable genotype-independent microbial diversity between animals, with the majority of taxa overlapping between CF and non-CF pairs. CF hepatic manifestations were variable, but included steatosis, necrosis, biliary hyperplasia, and biliary fibrosis. Gallbladder cystic mucosal hyperplasia was commonly found in 67% of CF animals. The majority of CF animals (85%) had pancreatic abnormalities, including extensive fibrosis, loss of exocrine pancreas, and islet disorganization. Interestingly, 2 of 13 CF animals retained predominantly normal pancreatic histology (84% to 94%) at time of death. Fecal elastase-1 levels from these CF animals were similar to non-CF controls, whereas all other CF animals evaluated were pancreatic insufficient (
- Published
- 2014
- Full Text
- View/download PDF
6. Lung Phenotype of Juvenile and Adult Cystic Fibrosis Transmembrane Conductance Regulator–Knockout Ferrets
- Author
-
Paul M. Kaminsky, Zoe A. Stewart, Ziying Yan, R. Marshall Pope, Nicholas W. Keiser, Xingshen Sun, Yaling Yi, John F. Engelhardt, J. Adam Goeken, Kalpaj R. Parekh, Danielle Fligg, Bo Liang, Hongshu Sui, Yi Song, Scott R. Tyler, Weihong Zhou, Thomas J. Lynch, Xiaoyan Wang, David K. Meyerholz, Xiaoming Liu, Joann M. Kinyon, Yulong Zhang, John T. Fisher, Weiliang Xie, Turan I.A. Evans, Alicia K. Olivier, and Timothy S. Frana
- Subjects
Pulmonary and Respiratory Medicine ,Pathology ,medicine.medical_specialty ,Cystic Fibrosis ,Mucociliary clearance ,Clinical Biochemistry ,Cystic Fibrosis Transmembrane Conductance Regulator ,Atelectasis ,Biology ,Air trapping ,Cystic fibrosis ,Animals, Genetically Modified ,medicine ,Animals ,Genetic Predisposition to Disease ,Lung ,Respiratory Tract Infections ,Molecular Biology ,Original Research ,Submucosal glands ,Age Factors ,Ferrets ,Cell Biology ,respiratory system ,medicine.disease ,Mucus ,Cystic fibrosis transmembrane conductance regulator ,Anti-Bacterial Agents ,respiratory tract diseases ,Intestines ,Disease Models, Animal ,Phenotype ,medicine.anatomical_structure ,Mucociliary Clearance ,Bacterial Translocation ,Disease Progression ,biology.protein ,medicine.symptom - Abstract
Chronic bacterial lung infections in cystic fibrosis (CF) are caused by defects in the CF transmembrane conductance regulator chloride channel. Previously, we described that newborn CF transmembrane conductance regulator–knockout ferrets rapidly develop lung infections within the first week of life. Here, we report a more slowly progressing lung bacterial colonization phenotype observed in juvenile to adult CF ferrets reared on a layered antibiotic regimen. Even on antibiotics, CF ferrets were still very susceptible to bacterial lung infection. The severity of lung histopathology ranged from mild to severe, and variably included mucus obstruction of the airways and submucosal glands, air trapping, atelectasis, bronchopneumonia, and interstitial pneumonia. In all CF lungs, significant numbers of bacteria were detected and impaired tracheal mucociliary clearance was observed. Although Streptococcus, Staphylococcus, and Enterococcus were observed most frequently in the lungs of CF animals, each animal displayed a predominant bacterial species that accounted for over 50% of the culturable bacteria, with no one bacterial taxon predominating in all animals. Matrix-assisted laser desorption–ionization time-of-flight mass spectrometry fingerprinting was used to quantify lung bacteria in 10 CF animals and demonstrated Streptococcus, Staphylococcus, Enterococcus, or Escherichia as the most abundant genera. Interestingly, there was significant overlap in the types of bacteria observed in the lung and intestine of a given CF animal, including bacterial taxa unique to the lung and gut of each CF animal analyzed. These findings demonstrate that CF ferrets develop lung disease during the juvenile and adult stages that is similar to patients with CF, and suggest that enteric bacterial flora may seed the lung of CF ferrets.
- Published
- 2014
- Full Text
- View/download PDF
7. Bioelectric Characterization of Epithelia from Neonatal CFTR Knockout Ferrets
- Author
-
Weihong Zhou, Hugo R. de Jonge, Xingshen Sun, Yaling Yi, Hongshu Sui, Ben J. Lee, Meihui Luo, Nicholas W. Keiser, Bo Liang, Weiliang Xie, Scott R. Tyler, Xiaoming Liu, John F. Engelhardt, Yulong Zhang, Yi Song, Kai Wang, John T. Fisher, and Gastroenterology & Hepatology
- Subjects
Pancreatic disease ,IBMX ,Cystic Fibrosis ,Phosphodiesterase Inhibitors ,Clinical Biochemistry ,Cystic Fibrosis Transmembrane Conductance Regulator ,Cystic fibrosis ,Membrane Potentials ,Animals, Genetically Modified ,chemistry.chemical_compound ,Gene Knockout Techniques ,Intestinal mucosa ,Cyclic AMP ,Electric Impedance ,Intestinal Mucosa ,Stomach ,Gallbladder ,Articles ,Hydrogen-Ion Concentration ,respiratory system ,Cystic fibrosis transmembrane conductance regulator ,medicine.anatomical_structure ,Phenotype ,Chloride channel ,Adenylyl Cyclases ,Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,Genotype ,Enzyme Activators ,Respiratory Mucosa ,Biology ,Chlorides ,SDG 3 - Good Health and Well-being ,Internal medicine ,Membrane Transport Modulators ,medicine ,Gastric mucosa ,Animals ,Molecular Biology ,Ion Transport ,Sodium ,Ferrets ,Epithelial Cells ,Cell Biology ,medicine.disease ,Enzyme Activation ,Disease Models, Animal ,Endocrinology ,chemistry ,Animals, Newborn ,Gastric Mucosa ,biology.protein - Abstract
Cystic fibrosis (CF) is a life-shortening, recessive, multiorgan genetic disorder caused by the loss of CF transmembrane conductance regulator (CFTR) chloride channel function found in many types of epithelia. Animal models that recapitulate the human disease phenotype are critical to understanding pathophysiology in CF and developing therapies. CFTR knockout ferrets manifest many of the phenotypes observed in the human disease, including lung infections, pancreatic disease and diabetes, liver disease, malnutrition, and meconium ileus. In the present study, we have characterized abnormalities in the bioelectric properties of the trachea, stomach, intestine, and gallbladder of newborn CF ferrets. Short-circuit current (I-SC) analysis of CF and wild-type (WT) tracheas revealed the following similarities and differences: (1) amiloride-sensitive sodium currents were similar between genotypes; (2) responses to 4,4'-diisothiocyano-2,2'-stilbene disulphonic acid were 3.3-fold greater in CF animals, suggesting elevated baseline chloride transport through non-CFTR channels in a subset of CF animals; and (3) a lack of 3-isobutyl-1-methylxanthine (IBMX)/forskolin-stimulated and N-(2-Naphthalenyl)-((3,5-dibromo-2,4-dihydroxyphenyl)methylene) glycine hydrazide (GlyH-101)-inhibited currents in CF animals due to the lack of CFTR. CFTR mRNA was present throughout all levels of the WT ferret and IBMX/forskolin-inducible I-SC was only observed in WT animals. However, despite the lack of CFTR function in the knockout ferret, the luminal pH of the CF ferret gallbladder, stomach, and intestines was not significantly changed relative to WT. The WT stomach and gallbladder exhibited significantly enhanced IBMX/forskolin I-SC responses and inhibition by GlyH-101 relative to CF samples. These findings demonstrate that multiple organs affected by disease in the CF ferret have bioelectric abnormalities consistent with the lack of cAMP-mediated chloride transport.
- Published
- 2013
- Full Text
- View/download PDF
8. Directing Integrin-linked Endocytosis of Recombinant AAV Enhances Productive FAK-dependent Transduction
- Author
-
Paul M. Kaminsky, Ziying Yan, Nicholas W. Keiser, John F. Engelhardt, and Diana C.M. Lei-Butters
- Subjects
viruses ,media_common.quotation_subject ,Genetic Vectors ,Integrin ,Eptifibatide ,Biology ,Endocytosis ,Focal adhesion ,Mice ,03 medical and health sciences ,Transduction (genetics) ,0302 clinical medicine ,Genes, Reporter ,Integrin complex ,Drug Discovery ,Genetics ,Animals ,Humans ,Luciferases ,Internalization ,Molecular Biology ,Cells, Cultured ,030304 developmental biology ,media_common ,Pharmacology ,Manganese ,0303 health sciences ,Gene Transfer Techniques ,Dependovirus ,Fibroblasts ,Vinculin ,Cell biology ,Focal Adhesion Protein-Tyrosine Kinases ,030220 oncology & carcinogenesis ,biology.protein ,Receptors, Virus ,Molecular Medicine ,Original Article ,Signal transduction ,Peptides ,Signal Transduction - Abstract
Recombinant adeno-associated virus (rAAV) is a widely used gene therapy vector. Although a wide range of rAAV serotypes can effectively enter most cell types, their transduction efficiencies (i.e., transgene expression) can vary widely depending on the target cell type. Integrins play important roles as coreceptors for rAAV infection, however, it remains unclear how integrin-dependent and -independent mechanisms of rAAV endocytosis influence the efficiency of intracellular virus processing and ultimately transgene expression. In this study, we examined the contribution of integrin-mediated endocytosis to transduction of fibroblasts by rAAV2. Mn(++)-induced integrin activation significantly enhanced (~17-fold) the efficiency of rAAV2 transduction, without altering viral binding or endocytosis. rAAV2 subcellular localization studies demonstrated that Mn(++) promotes increased clustering of rAAV2 on integrins and recruitment of intracellular vinculin (an integrin effector) to sites of rAAV2 binding at the cell surface. Focal adhesion kinase (FAK), a downstream effector of integrin signals, was essential for rAAV2/integrin complex internalization and transduction. These findings support a model whereby integrin activation at the cell surface can redirect rAAV2 toward a FAK-dependent entry pathway that is more productive for cellular transduction. This pathway appears to be conserved for other rAAV serotypes that contain a capsid integrin-binding domain (AAV1 and AAV6).
- Published
- 2012
- Full Text
- View/download PDF
9. Defective innate immunity and hyperinflammation in newborn cystic fibrosis transmembrane conductance regulator-knockout ferret lungs
- Author
-
Elizabeth Stroebele, Xingshen Sun, Susan E. Birket, Weihong Zhou, Mark J. Stevens, Idil A. Evans, Guillermo J. Tearney, Joseph R. Nellis, J. Kirk Harris, Steven M. Rowe, Adrianne K. Crooke, Nicholas W. Keiser, John F. Engelhardt, Kengyeh K. Chu, and Scott R. Tyler
- Subjects
Pulmonary and Respiratory Medicine ,Cystic Fibrosis ,Proteome ,Mucociliary clearance ,Clinical Biochemistry ,Cystic Fibrosis Transmembrane Conductance Regulator ,Inflammation ,Cystic fibrosis ,Gene Knockout Techniques ,medicine ,Animals ,Pseudomonas Infections ,Molecular Biology ,Original Research ,Submucosal glands ,Innate immune system ,Lung ,biology ,medicine.diagnostic_test ,Ferrets ,Cell Biology ,respiratory system ,medicine.disease ,Cystic fibrosis transmembrane conductance regulator ,Immunity, Innate ,respiratory tract diseases ,Trachea ,medicine.anatomical_structure ,Bronchoalveolar lavage ,Animals, Newborn ,Mucociliary Clearance ,Immunology ,Pseudomonas aeruginosa ,biology.protein ,Cytokines ,medicine.symptom ,Inflammation Mediators - Abstract
Mucociliary clearance (MCC) and submucosal glands are major components of airway innate immunity that have impaired function in cystic fibrosis (CF). Although both of these defense systems develop postnatally in the ferret, the lungs of newborn ferrets remain sterile in the presence of a functioning cystic fibrosis transmembrane conductance regulator gene. We evaluated several components of airway innate immunity and inflammation in the early CF ferret lung. At birth, the rates of MCC did not differ between CF and non-CF animals, but the height of the airway surface liquid was significantly reduced in CF newborn ferrets. CF ferrets had impaired MCC after 7 days of age, despite normal rates of ciliogenesis. Only non-CF ferrets eradicated Pseudomonas directly introduced into the lung after birth, whereas both genotypes could eradicate Staphylococcus. CF bronchoalveolar lavage fluid (BALF) had significantly lower antimicrobial activity selectively against Pseudomonas than non-CF BALF, which was insensitive to changes in pH and bicarbonate. Liquid chromatography–tandem mass spectrometry and cytokine analysis of BALF from sterile Caesarean-sectioned and nonsterile naturally born animals demonstrated CF-associated disturbances in IL-8, TNF-α, and IL-β, and pathways that control immunity and inflammation, including the complement system, macrophage functions, mammalian target of rapamycin signaling, and eukaryotic initiation factor 2 signaling. Interestingly, during the birth transition, IL-8 was selectively induced in CF BALF, despite no genotypic difference in bacterial load shortly after birth. These results suggest that newborn CF ferrets have defects in both innate immunity and inflammatory signaling that may be important in the early onset and progression of lung disease in these animals.
- Published
- 2014
10. Inhibition of Human Immunodeficiency Virus Type 1 (HIV-1) Replication by HIV-1-Based Lentivirus Vectors Expressing Transdominant Rev
- Author
-
Richard A. Morgan, Mario R. Mautino, and Nicholas W. Keiser
- Subjects
T-Lymphocytes ,viruses ,Genetic Vectors ,Immunology ,HIV Core Protein p24 ,Gene Expression ,Virus Replication ,Microbiology ,Virus ,Cell Line ,Transduction (genetics) ,Plasmid ,Retrovirus ,Proviruses ,Simian retrovirus ,Transduction, Genetic ,Virology ,Murine leukemia virus ,Humans ,Vector (molecular biology) ,biology ,Virus Assembly ,rev Gene Products, Human Immunodeficiency Virus ,Genetic Therapy ,Gene Therapy ,biology.organism_classification ,Fusion Proteins, gag-pol ,Blotting, Southern ,Gene Products, rev ,Viral replication ,Insect Science ,HIV-1 ,Moloney murine leukemia virus - Abstract
Retrovirus vectors expressing transdominant-negative mutants of Rev (TdRev) inhibit human immunodeficiency virus type 1 (HIV-1) replication by preventing the nuclear export of unspliced viral transcripts, thus inhibiting the synthesis of Gag-Pol, Env, and genomic RNA. The use of HIV-1–based vectors to express TdRev would have the advantage of allowing access to nondividing hematopoietic cells. It would also provide additional levels of protection by sequestering the viral regulatory proteins Tat and Rev, competing for encapsidation into wild-type virions, and inhibiting reverse transcription. Here we describe HIV-1-based vectors that express TdRev. These vectors contain mutations in the splicing signals or replacement of the Rev-responsive element by the simian retrovirus type 1 constitutive transport element, making them less sensitive to the inhibitory effects of TdRev. In addition, overexpression of Rev and the use of an HIV-1 helper plasmid that drives high levels of Gag-Pol synthesis were used to transiently overcome the inhibition by TdRev of the synthesis of Gag-Pol during vector production. SupT1 cells transduced with these vectors were more resistant to HIV-1 replication than cells transduced with Moloney murine leukemia virus-based vectors expressing TdRev. Furthermore, we show that these vectors can be mobilized by the wild-type virus, reducing the infectivity of virions escaping inhibition and conferring protection against HIV-1 replication to previously untransduced cells.
- Published
- 2001
- Full Text
- View/download PDF
11. Activation of MHC Class I, II, and CD40 Gene Expression by Histone Deacetylase Inhibitors
- Author
-
Geoffrey Catalano, Michelle A. Romano, Carleton C. Stewart, Frank Santaniello, Nicholas W. Keiser, Thomas B. Tomasi, William J. Magner, Catherine Grande, and A. Latif Kazim
- Subjects
Genes, MHC Class II ,Immunology ,Genes, MHC Class I ,Apoptosis ,Hydroxamic Acids ,Interferon-gamma ,Mice ,Gene expression ,MHC class I ,Tumor Cells, Cultured ,Animals ,Humans ,Immunology and Allergy ,RNA, Messenger ,Epigenetics ,CD40 Antigens ,Enzyme Inhibitors ,Regulation of gene expression ,biology ,Cell Cycle ,Histocompatibility Antigens Class II ,Nuclear Proteins ,Chromatin ,Histone Deacetylase Inhibitors ,Histone ,Gene Expression Regulation ,Acetylation ,Trans-Activators ,biology.protein ,Cancer research ,Histone deacetylase - Abstract
Epigenetic mechanisms are involved in regulating chromatin structure and gene expression through repression. In this study, we show that histone deacetylase inhibitors (DAIs) that alter the acetylation of histones in chromatin enhance the expression of several genes on tumor cells including: MHC class I, II, and the costimulatory molecule CD40. Enhanced transcription results in a significant increase in protein expression on the tumor cell surface, and expression can be elicited on some tumors that are unresponsive to IFN-γ. The magnitude of induction of these genes cannot be explained by the effect of DAIs on the cell cycle or enhanced apoptosis. Induction of class II genes by DAIs was accompanied by activation of a repressed class II transactivator gene in a plasma cell tumor but, in several other tumor cell lines, class II was induced in the apparent absence of class II transactivator transcripts. These findings also suggest that the abnormalities observed in some tumors in the expression of genes critical to tumor immunity may result from epigenetic alterations in chromatin and gene regulation in addition to well-established mutational mechanisms.
- Published
- 2000
- Full Text
- View/download PDF
12. A Novel Chimeric Adenoassociated Virus 2/Human Bocavirus 1 Parvovirus Vector Efficiently Transduces Human Airway Epithelia
- Author
-
Nicholas W. Keiser, John F. Engelhardt, Fang Cheng, Xuefeng Deng, Yi Song, Jianming Qiu, and Ziying Yan
- Subjects
Pharmacology ,0303 health sciences ,biology ,Genetic enhancement ,viruses ,Human bocavirus ,Apical membrane ,biology.organism_classification ,Virology ,Cystic fibrosis transmembrane conductance regulator ,Virus ,Viral vector ,03 medical and health sciences ,Transduction (genetics) ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Drug Discovery ,biology.protein ,Genetics ,Molecular Medicine ,Original Article ,Gene ,Molecular Biology ,030304 developmental biology - Abstract
Human bocavirus virus-1 (HBoV1), a newly discovered autonomous parvovirus with a 5,500 nt genome, efficiently infects human-polarized airway epithelia (HAE) from the apical membrane. We hypothesized that the larger genome and high airway tropism of HBoV1 would be ideal for creating a viral vector for lung gene therapy. To this end, we successfully generated recombinant HBoV1 (rHBoV1) from an open reading frames–disrupted rHBoV1 genome that efficiently transduces HAE from the apical surface. We next evaluated whether HBoV1 capsids could package oversized rAAV2 genomes. These studies created a rAAV2/HBoV1 chimeric virus (5.5 kb genome) capable of apically transducing HAE at 5.6- and 70-fold greater efficiency than rAAV1 or rAAV2 (4.7-kb genomes), respectively. Molecular studies demonstrated that viral uptake from the apical surface was significantly greater for rAAV2/HBoV1 than for rAAV2 or rAAV1, and that polarization of airway epithelial cells was required for HBoV1 capsid–mediated gene transfer. Furthermore, rAAV2/HBoV1-CFTR virus containing the full-length cystic fibrosis transmembrane conductance regulator (CFTR) gene coding sequence and the strong CBA promoter efficiently corrected CFTR-dependent chloride transport in cystic fibrosis (CF) HAE. In summary, using the combined advantages of AAV and HBoV1, we have developed a novel and promising viral vector for CF lung gene therapy and also potentially HBoV1 vaccine development.
- Published
- 2013
13. Gene Delivery to the Airway
- Author
-
Nicholas W. Keiser and John F. Engelhardt
- Subjects
Genetic enhancement ,Transgene ,Cell Culture Techniques ,Gene Expression ,Respiratory Mucosa ,Gene delivery ,Biology ,Article ,Transduction (genetics) ,Transduction, Genetic ,Genetics ,medicine ,Animals ,Humans ,Transgenes ,Genetics (clinical) ,Cell Line, Transformed ,Goblet cell ,Reporter gene ,Gene Transfer Techniques ,Epithelial Cells ,respiratory system ,Cell biology ,medicine.anatomical_structure ,Models, Animal ,Immunology ,Heterografts ,Respiratory epithelium ,Stem cell - Abstract
Model systems of differentiated airway epithelium have played a significant role in research pertaining to airway biology, pathophysiology, and gene therapy. The success of such systems is dependent on the ability to reconstitute the native cellular composition and architecture of the airway, within a setting that retains adequate flexibility for experimental manipulation. Furthermore, human airway models have provided significant advantages over other models, since the cell biology of the airway epithelium of humans can differ substantially in function and cellular composition from that of other species such as mice and rats. For example, the predominant secretory cell type in humans is the goblet cell, whereas in mice and rats it is the Clara and serous cell, respectively. Another important consideration is the marked variation observed in the tropism of recombinant viruses commonly used for gene therapy (e.g., adeno-associated virus) with recipient cells from different species. One of the most widely used human systems to date consists of polarized monolayers of primary airway epithelial cells grown on permeable membrane supports (Karp et al., 2002; Randell et al., 2011; Yamaya et al., 1992). For many studies, this system has provided adequate differentiation when cells are grown at the air-liquid interface. However, the extent of mucociliary differentiation in this experimental model is often inadequate for studies related to in vivo airway epithelial functions. To circumvent these limitations of current airway models, tracheal xenograft models have been developed to study gene transfer and airway pathophysiology in human genetic diseases (Wilson, 1997). These airway xenograft models have proved extremely useful in studying host cell–vector interactions (Engelhardt et al., 1993b; Engelhardt et al., 1992; Goldman and Wilson, 1995) with human airway epithelium, as well as pathophysiology and gene therapy of the cystic fibrosis airways (Goldman et al., 1997; Zhang et al., 1995; Zhang and Engelhardt, 1999; Zhang et al., 1998; Zhang et al., 1996), and the identification of progenitor/stem cell targets for gene therapy in the human airway (Duan et al., 1998a; Engelhardt et al., 1995). This unit describes generation of and gene transfer to several commonly used airway models. Isolation (see Basic Protocol 1) and transduction (see Basic Protocol 2) of primary airway epithelial cells are first described. Next, the preparation of polarized airway epithelial monolayers is outlined (see Basic Protocol 3). Transduction of these polarized cells by recombinant adenovirus, adeno-associated virus, retrovirus, or lentivirus is also described (see Basic Protocol 4). Methods are presented for generation of human and ferret tracheal xenografts (see Basic Protocol 5) as well as both ex vivo and in vivo gene transfer to these xenografts (see Basic Protocol 6). Finally, a method for in vivo gene delivery to the lungs of rodents is included (see Basic Protocol 7). Some methods for the evaluation of transgene expression are given in the support protocols. A method for harvesting xenografts for morphological analysis is described (see Support Protocol 1). The reporter gene β-galactosidase can be detected either histochemically (Support Protocol 2) or immunocytochemically (Support Protocol 3). If green fluorescent protein (GFP) is used as a reporter gene, it can be detected fluorescently (see Support Protocol 4). Finally, histochemical detection of alkaline phosphatase gene activity is described (see Support Protocol 5). CAUTION: Radioactive, biological, and chemical substances require special handling; see appendix 2a for guidelines.
- Published
- 2013
- Full Text
- View/download PDF
14. Abnormal endocrine pancreas function at birth in cystic fibrosis ferrets
- Author
-
Nicholas W. Keiser, Ziying Yan, Diana Lei, John F. Engelhardt, Hongshu Sui, Alicia K. Olivier, Zoe A. Stewart, Weihong Zhou, Kai Wang, Guiying Li, Weiliang Xie, Turan I.A. Evans, Xingshen Sun, Yaling Yi, Bo Liang, Andrew W. Norris, John T. Fisher, David K. Meyerholz, and Shanming Hu
- Subjects
Male ,medicine.medical_specialty ,Cystic Fibrosis ,medicine.medical_treatment ,Apoptosis ,Biology ,Glucagon ,Cystic fibrosis ,Diabetes mellitus genetics ,Gene Knockout Techniques ,Islets of Langerhans ,Species Specificity ,Internal medicine ,Diabetes mellitus ,Glucose Intolerance ,Insulin Secretion ,medicine ,Diabetes Mellitus ,Endocrine system ,Animals ,Insulin ,Cells, Cultured ,Ferrets ,Pancreatic Ducts ,General Medicine ,medicine.disease ,Fibrosis ,Pancreas, Exocrine ,Disease Models, Animal ,medicine.anatomical_structure ,Endocrinology ,Glucose ,Technical Advance ,Animals, Newborn ,Pancreatitis ,Hyperglycemia ,Disease Progression ,Female ,Pancreas ,Dilatation, Pathologic - Abstract
Diabetes is a common comorbidity in cystic fibrosis (CF) that worsens prognosis. The lack of an animal model for CF-related diabetes (CFRD) has made it difficult to dissect how the onset of pancreatic pathology influences the emergence of CFRD. We evaluated the structure and function of the neonatal CF endocrine pancreas using a new CFTR-knockout ferret model. Although CF kits are born with only mild exocrine pancreas disease, progressive exocrine and endocrine pancreatic loss during the first months of life was associated with pancreatic inflammation, spontaneous hyperglycemia, and glucose intolerance. Interestingly, prior to major exocrine pancreas disease, CF kits demonstrated significant abnormalities in blood glucose and insulin regulation, including diminished first-phase and accentuated peak insulin secretion in response to glucose, elevated peak glucose levels following glucose challenge, and variably elevated insulin and C-peptide levels in the nonfasted state. Although there was no difference in lobular insulin and glucagon expression between genotypes at birth, significant alterations in the frequencies of small and large islets were observed. Newborn cultured CF islets demonstrated dysregulated glucose-dependent insulin secretion in comparison to controls, suggesting intrinsic abnormalities in CF islets. These findings demonstrate that early abnormalities exist in the regulation of insulin secretion by the CF endocrine pancreas.
- Published
- 2012
15. Distinct transduction difference between adeno-associated virus type 1 and type 6 vectors in human polarized airway epithelia
- Author
-
Diana C.M. Lei-Butters, Ziying Yan, Nicholas W. Keiser, and John F. Engelhardt
- Subjects
Mutant ,Genetic Vectors ,Respiratory System ,Biology ,medicine.disease_cause ,Article ,Transduction Polarity ,Cell Line ,03 medical and health sciences ,Transduction (genetics) ,AAV1 and AAV6 ,0302 clinical medicine ,Species Specificity ,Transduction, Genetic ,Cell polarity ,Genetics ,medicine ,Human Airway Epithelia ,Humans ,Amino Acids ,Serotyping ,Molecular Biology ,Adeno-associated virus ,Cells, Cultured ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Wild type ,Cell Polarity ,Epithelial Cells ,Dependovirus ,Molecular biology ,Amino acid ,Cell biology ,HEK293 Cells ,chemistry ,Capsid ,Cell culture ,030220 oncology & carcinogenesis ,Mutation ,Molecular Medicine ,Capsid Proteins ,HeLa Cells - Abstract
Of the many biologically isolated adeno-associated virus (AAV) serotypes, AAV1 and AAV6 share the highest degree of sequence homology, with only six different capsid residues. We compared the transduction efficiencies of rAAV1 and rAAV6 in primary polarized human airway epithelia and found significant differences in their abilities to transduce epithelia from the apical and basolateral membranes. rAAV1 transduction was ~10-fold higher than rAAV6 following apical infection, whereas rAAV6 transduction was ~10-fold higher than rAAV1 following basolateral infection. Furthermore, rAAV6 demonstrated significant polarity of transduction (100-fold; basolateral » apical), whereas rAAV1 transduced from both membranes with equal efficiency. To evaluate capsid residues responsible for the observed serotype differences, we mutated the six divergent amino acids either alone or in combination. Results from these studies demonstrated that capsid residues 418 and 531 most significantly controlled membrane polarity differences in transduction between serotypes, with the rAAV6-D418E/K531E mutant demonstrating decreased (~10-fold) basolateral transduction and the rAAV1-E418D/E531K mutant demonstrating a transduction polarity identical to rAAV6-WT (wild type). However, none of the rAAV6 mutants obtained apical transduction efficiencies of rAAV1-WT, suggesting that all six divergent capsid residues in AAV1 act in concert to improve apical transduction of HAE.
- Published
- 2012
16. New animal models of cystic fibrosis: what are they teaching us?
- Author
-
Nicholas W. Keiser and John F. Engelhardt
- Subjects
Pulmonary and Respiratory Medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Pathology ,medicine.medical_specialty ,Cystic Fibrosis ,Swine ,Cystic Fibrosis Transmembrane Conductance Regulator ,Disease ,Cystic fibrosis ,Article ,Mice ,Conditional gene knockout ,medicine ,Animals ,Humans ,Lung ,Pancreas ,biology ,business.industry ,Ferrets ,medicine.disease ,Cystic fibrosis transmembrane conductance regulator ,Pathophysiology ,Disease Models, Animal ,medicine.anatomical_structure ,Knockout mouse ,biology.protein ,business - Abstract
Purpose of review Cystic fibrosis is the first human genetic disease to benefit from the directed engineering of three different species of animal models (mice, pigs, and ferrets). Recent studies on the cystic fibrosis pig and ferret models are providing new information about the pathophysiology of cystic fibrosis in various organ systems. Additionally, new conditional cystic fibrosis transmembrane conductance regulator (CFTR) knockout mice are teaching unexpected lessons about CFTR function in surprising cellular locations. Comparisons between these animal models and the human condition are key to dissecting the complexities of disease pathophysiology in cystic fibrosis. Recent findings Cystic fibrosis pigs and ferrets have provided new models to study the spontaneous development of disease in the lung and pancreas, two organs that are largely spared overt spontaneous disease in cystic fibrosis mice. New cystic fibrosis mouse models are now interrogating CFTR functions involved in growth and inflammation at an organ-based level using conditional knockout technology. Together, these models are providing new insights on the human condition. Summary Basic and clinical cystic fibrosis research will benefit greatly from the comparative pathophysiology of cystic fibrosis mice, pigs, and ferrets. Both similarities and differences between these three cystic fibrosis models will inform pathophysiologically important mechanisms of CFTR function in humans and aid in the development of both organ-specific and general therapies for cystic fibrosis.
- Published
- 2011
17. Unique Characteristics of AAV1, 2, and 5 Viral Entry, Intracellular Trafficking, and Nuclear Import Define Transduction Efficiency in HeLa Cells
- Author
-
Nicholas W. Keiser, John F. Engelhardt, Diana C.M. Lei-Butters, Yulong Zhang, and Ziying Yan
- Subjects
viruses ,Cell ,Genetic Vectors ,Active Transport, Cell Nucleus ,Biology ,HeLa ,Cell membrane ,Transduction (genetics) ,Viral entry ,Transduction, Genetic ,Genetics ,medicine ,Humans ,Molecular Biology ,Research Articles ,Cell Nucleus ,Cell Membrane ,Dependovirus ,biology.organism_classification ,Virology ,Cell biology ,Cell nucleus ,medicine.anatomical_structure ,Molecular Medicine ,Nuclear transport ,Intracellular ,HeLa Cells - Abstract
Biological differences between recombinant adeno-associated virus (rAAV) serotypes define their efficiencies in expressing a transgene in a particular target cell. Few studies have directly compared how differences in viral entry, intracellular trafficking, and nuclear import of rAAV serotypes influence the effectiveness of transduction in the same cell type. We evaluated these characteristics for three rAAV serotypes in HeLa cells, using biochemical techniques and fluorescence-based detection of multiple serotypes in the same cell. Although rAAV2 exhibited the slowest entry, intracellular trafficking, and nuclear import among the three serotypes, it elicited the highest levels of transduction. Conversely, rAAV1 exhibited more rapid entry and nuclear import than the other serotypes, yet was ineffective at transducing HeLa cells due to impaired capsid disassembly in the nucleus. rAAV5, which entered the cell less rapidly than rAAV1, was imported efficiently into the nucleus, but then rapidly degraded, resulting in poor transduction of HeLa cells. We conclude that rAAV1, 2, and 5 utilize distinct mechanisms for intracellular trafficking, and that post-nuclear events play an important role in determining the efficiency of HeLa cell transduction by these serotypes. Thus, overcoming post-nuclear barriers that limit uncoating and/or promote virion degradation may enhance the efficiency of certain AAV serotypes.
- Published
- 2011
18. Spatial and temporal expression patterns of the choroideremia gene in the mouse retina
- Author
-
Nicholas W, Keiser, Waixing, Tang, Zhangyong, Wei, and Jean, Bennett
- Subjects
Mice, Inbred ICR ,Alkyl and Aryl Transferases ,Blotting, Western ,Gene Expression ,RNA Probes ,Retina ,Immunoenzyme Techniques ,Mice ,Animals ,RNA, Messenger ,Rabbits ,Fluorescent Antibody Technique, Indirect ,Choroideremia ,In Situ Hybridization ,Adaptor Proteins, Signal Transducing - Abstract
Choroideremia (CHM), an X-linked retinal disease, is caused by mutations affecting the CHM gene. This gene encodes REP-1, which functions in the covalent modifications of proteins involved in vesicle trafficking. The disease affects several cell types in the retina, but it is not known which cell types contribute directly or indirectly to disease progression. A study of the expression patterns of Chm and the related gene Chml in the mouse retina was undertaken in order to address this issue.The expression patterns of Chm and Chml were determined by in situ hybridization. The localization of the Chm protein product, Rep-1, was determined spatially and temporally in the mouse retina by immunohistochemistry.Chm and Chml mRNA were found in every major layer of the retina in adult mice. During development, Rep-1 protein localization changes from a fairly diffuse pattern during embryogenesis to a more specific pattern at the time of retinal differentiation. In adulthood, Rep-1 localizes to distinct cellular compartments in multiple retinal cell types.Chm and Chml have the same broad expression profile in the mouse retina. In particular, the Chm transcript and corresponding protein are found in cell types other than those thought to be primarily affected in the human disease. These results have important implications for approaches with which to develop a relevant mouse model of choroideremia and for therapeutic strategies for this disease.
- Published
- 2005
19. Improved titers of HIV-based lentiviral vectors using the SRV-1 constitutive transport element
- Author
-
Nicholas W. Keiser, Mario R. Mautino, and Richard A. Morgan
- Subjects
Acquired Immunodeficiency Syndrome ,biology ,Genetic enhancement ,Genetic Vectors ,Mutagenesis (molecular biology technique) ,rev Gene Products, Human Immunodeficiency Virus ,Genetic Therapy ,biology.organism_classification ,Virology ,Viral vector ,Retroviruses, Simian ,Titer ,Gene Products, rev ,Gene expression ,Lentivirus ,Genetics ,Mutagenesis, Site-Directed ,Molecular Medicine ,Humans ,Vector (molecular biology) ,Molecular Biology ,Gene - Abstract
The development of lentiviral vectors that use Rev-independent mechanisms of nuclear export for their genomic RNA could facilitate the construction of novel anti-HIV vectors. We have improved the titers of Rev-independent lentiviral vectors having the SRV-1 CTE by mutating the major splice donor and acceptor sites present in the vector and by relocalization of the CTE sequences adjacent to the HIV-1 3'LTR. These two modifications have additive beneficial effects on vector titers and packaging efficiency. Packaging these CTE+ vectors expressing marker genes with a Rev-dependent HIV-1 helper vector yields higher titers than are obtained using a Rev-dependent lentiviral vector.
- Published
- 2000
20. 135. AAV-Mediated Gene Delivery To Evaluate the Biology of an Inherited Macular Degeneration
- Author
-
Nicholas W. Keiser, Waixing Tang, Jean Bennett, Zhanyong Wei, Albert M. Maguire, Jeannette L. Bennicelli, and Daniel C. Chung
- Subjects
Pathology ,medicine.medical_specialty ,genetic structures ,Population ,Biology ,Drusen ,Extracellular matrix protein 1 ,Degenerative disease ,Drug Discovery ,Genetics ,medicine ,education ,Molecular Biology ,Pharmacology ,Retina ,education.field_of_study ,Retinal pigment epithelium ,Anatomy ,Macular degeneration ,medicine.disease ,eye diseases ,medicine.anatomical_structure ,Choroidal neovascularization ,Molecular Medicine ,sense organs ,medicine.symptom - Abstract
Malattia Leventinese (ML) is an autosomal dominant inherited macular degeneration with a middle-age onset. Phenotypically, the disease closely resembles Age-related Macular Degeneration (AMD), a condition affecting 20% of the population over age 65 and the leading cause of blindness in the elderly in the Western world. There is no cure for macular degeneration, and the supportive treatments are limited. Patients with ML suffer from decreased visual acuity and have a characteristic radial pattern of spots, called drusen, in their maculas. Clinical exam reveals geographic atrophy, pigmentary changes, and choroidal neovascularization. Histopathology reveals deposits between the retinal pigment epithelium (RPE) and Bruch's membrane, similar to what is found in AMD. A missense mutation, R345W, in the Epidermal Growth Factor Containing Fibrillin-like Extracellular Matrix Protein 1 (EFEMP1) gene is responsible for the disease, and leads to the misfolding and inefficient secretion of the protein. To gain insight into the role of EFEMP1 in the retina in health and disease, we studied the localization of the wild-type Efemp1 protein and mRNA in the developing mouse retina. Efemp1 protein localizes first to the inner retina and RPE of the mouse, with localization to photoreceptors detectable by postnatal day 14. We also found that Efemp1 protein colocalized with TIMP-3 in the inner retina throughout murine ocular development. Efemp1 mRNA was broadly distributed across the developing retina, and then was concentrated in photoreceptors and ganglion cells by adulthood. An AAV2/5 vector carrying FLAG-tagged mutant Efemp1 was delivered into the wild-type mouse retina, and this resulted in the targeting of the fusion protein to the photoreceptors and RPE. There, it was detected in cytoplasmic aggregates by EM immunohistochemistry. We also identified radial lesions resembling those seen in ML patients. Studies involving non-human primates are in progress. Our results demonstrate that EFEMP1 is a component of the inner and outer retina, but that the mutant form aggregates in the cytoplasm and induces lesions by a yet unknown mechanism. Generation of ML-like lesions in mice and non-human primates via gene delivery promises to be useful in generating animal models for macular degenerative disease in order to define the pathogenetics of the disease and to identify therapeutic targets.
- Published
- 2005
- Full Text
- View/download PDF
21. 886. AAV-Mediated Somatic Gene Transfer as an Approach To Delineate Pathogenic Mechanisms in an Autosomal Dominant Blindness Disorder Resembling Age-Related Macular Degeneration (AMD)
- Author
-
Jean Bennett, Nicholas W. Keiser, Arvydas Maminishkis, Sheldon S. Miller, and Jeannette L. Bennicelli
- Subjects
Pharmacology ,Genetics ,education.field_of_study ,Retinal pigment epithelium ,Population ,Biology ,Drusen ,Macular degeneration ,medicine.disease ,eye diseases ,Cell biology ,Extracellular matrix protein 1 ,medicine.anatomical_structure ,Cell culture ,Drug Discovery ,Cell polarity ,medicine ,Molecular Medicine ,Secretion ,sense organs ,education ,Molecular Biology - Abstract
Malattia Leventinese (ML) is an autosomal dominant inherited macular degeneration with a middle-age onset. Phenotypically, the disease closely resembles Age-related Macular Degeneration (AMD), a condition affecting 20% of the population over age 65 and the leading cause of blindness in the elderly in the Western world. Patients with ML suffer from decreased visual acuity and have a characteristic radial pattern of deposits, called drusen, which form beneath the retinal pigment epithelium (RPE). A single missense mutation (R345W) in the Epidermal Growth Factor Containing Fibrillin-like Extracellular Matrix Protein 1 (EFEMP1) gene is responsible for the disease, and leads to the misfolding and inefficient secretion of the protein. In normal retinas, the protein localizes to the inner photoreceptor matrix (IPM), apical to the RPE, while in individuals with ML, it resides between the RPE and drusen. We therefore surmised that wild-type EFEMP1 is apically secreted by the RPE, and that the R345W mutation misdirects this secretion basolaterally. To test this hypothesis, we utilized a cell culture system consisting of polarized monolayers of human fetal RPE (hfRPE) cells grown on Transwell membranes. AAV2/1 vectors containing FLAG-tagged wild-type or mutant EFEMP1 were used to transduce the hfRPE cells with high efficiency. Transduction did not affect hfRPE cell polarization or tight junction formation, as shown by immunofluorescence staining for Na+/K+ ATPase, CD147, and ZO-1. Immunoprecipitation of EFEMP1-FLAG from the basolateral and apical medium revealed that, contrary to our hypothesis, EFEMP1 is secreted basolaterally, and that this polarized secretion pattern is not altered by the R345W mutation. In addition, we found that this pattern of secretion was distinct from dog epithelial (MDCK) cells, which secreted the protein apically. Additional immunoprecipitation and immunofluorescence experiments indicated that the R345W mutation resulted in retention of EFEMP1 within polarized RPE cells. We conclude that EFEMP1 is directionally secreted from epithelial cells, and that its basolateral secretion from RPE cells may be indicative of some yet unknown function in the extracellular matrix underlying those cells, within the local environment of drusen formation in ML patients. However, the absence of an alteration in polarized secretion suggests that the intracellular retention of mutant EFEMP1 may be a major contributing factor to the progression of ML.
- Published
- 2006
- Full Text
- View/download PDF
22. Unique Characteristics of AAV1, 2, and 5 Viral Entry, Intracellular Trafficking, and Nuclear Import Define Transduction Efficiency in HeLa Cells.
- Author
-
Nicholas W. Keiser, Ziying Yan, Yulong Zhang, Diana C.M. Lei-Butters, and John F. Engelhardt
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.