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










Publication year range
1.
Commun Med (Lond) ; 2(1): 148, 2022 Nov 21.
Article in English | MEDLINE | ID: mdl-36414773

ABSTRACT

BACKGROUND: Adeno-associated virus (AAV) vectors are stored and shipped frozen which poses logistic and economic barriers for global access to these therapeutics. To address this issue, we developed a method to stabilize AAV serotype 9 (AAV9) in a film matrix that can be stored at ambient temperature and administered by systemic injection. METHODS: AAV9 expressing the luciferase transgene was mixed with formulations, poured into molds and films dried under aseptic conditions. Films were packaged in individual particle-free bags with foil overlays and stored at various temperatures under controlled humidity. Recovery of AAV9 from films was determined by serial dilution of rehydrated film in media and infection of HeLa RC32 cells. Luciferase expression was compared to that of films rehydrated immediately after drying. Biodistribution of vector was determined by in vivo imaging and quantitative real-time PCR. Residual moisture in films was determined by Karl Fischer titration. RESULTS: AAV9 embedded within a film matrix and stored at 4 °C for 5 months retained 100% of initial titer. High and low viscosity formulations maintained 90 and 85% of initial titer after 6 months at 25 °C respectively. AAV was not detected after 4 months in a Standard Control Formulation under the same conditions. Biodistribution and transgene expression of AAV stored in film at 25 or 4 °C were as robust as vector stored at -80 °C in a Standard Control Formulation. CONCLUSIONS: These results suggest that storage of AAV in a film matrix facilitates easy transport of vector to remote sites without compromising in vivo performance.


Adeno-associated viruses (AAVs) are small viruses that are used to deliver medicines and vaccines. Prior to administration, they are stored in freezers set to very low temperatures and must be discarded if they thaw during transportation to clinics. AAV was embedded in a film to protect the virus during transportation and storage. The virus remained stable for 6 months at room temperature and during shipment from Texas to North Carolina. The ability to store and transport AAV without the need for complex packaging and temperature control will increase global access to vaccines and other medicines that use AAVs for delivery.

2.
Hum Gene Ther ; 32(23-24): 1450-1456, 2021 12.
Article in English | MEDLINE | ID: mdl-34415793

ABSTRACT

Infants and older adults are especially vulnerable to infection by respiratory syncytial virus (RSV), which can cause significant illness and irreparable damage to the lower respiratory tract and for which an effective vaccine is not readily available. Palivizumab, a recombinant monoclonal antibody (mAb), is an approved therapeutic for RSV infection for use in high-risk infants only. Due to several logistical issues, including cost of goods and scale-up limitations, palivizumab is not approved for other populations that are vulnerable to severe RSV infections, such as older adults. In this study, we demonstrate that intranasal delivery of adeno-associated virus serotype 9 (AAV9) vector expressing palivizumab or motavizumab, a second-generation version of palivizumab, significantly reduced the viral load in the lungs of the BALB/c mouse model of RSV infection. Notably, we demonstrate that AAV9 vector-mediated prophylaxis against RSV was effective despite the presence of serum-circulating neutralizing AAV9 antibodies. These findings substantiate the feasibility of repeatedly administering AAV9 vector to the airway for seasonal prophylaxis against RSV, thereby expanding the application of vectored delivery of mAbs as an effective prophylaxis strategy against various airborne viruses.


Subject(s)
Dependovirus , Respiratory Syncytial Virus Infections , Animals , Antiviral Agents , Dependovirus/genetics , Lung , Mice , Mice, Inbred BALB C , Palivizumab/therapeutic use , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/prevention & control
3.
Mol Ther Methods Clin Dev ; 22: 1-10, 2021 Sep 10.
Article in English | MEDLINE | ID: mdl-34258325

ABSTRACT

Gene therapy for hypercholesterolemia offers the potential to sustainably ameliorate disease for life with a single dose. In this study, we demonstrate the combinatorial effects of codon and vector optimization, which significantly improve the efficacy of an adeno-associated virus (AAV) vector in the low-density lipoprotein receptor (LDLR)-deficient mouse model (Ldlr -/-, Apobec1 -/- double knockout [DKO]). This study investigated vector efficacy following the combination of intervening sequence 2 (IVS2) of the human beta-globin gene and codon optimization with the previously developed gain-of-function, human LDLR triple-mutant variant (hLDLR-L318D/K809R/C818A) in the treatment of homozygous familial hypercholesterolemia (HoFH). Vector doses as low as 3 × 1011 genome copies (GC)/kg achieved a robust reduction of serum low-density lipoprotein cholesterol (LDL-C) by 98% in male LDLR-deficient mice. Less efficient LDL-C reduction was observed in female mice, which was attributable to lower gene transfer efficiency in liver. We also observed persistent and stable transgene expression for 120 days, with LDL-C levels being undetectable in male DKO mice treated with the second-generation vector. In conclusion, codon and vector optimization enhanced transgene expression and reduced serum LDL-C levels effectively at a lower dose in LDLR-deficient mice. The second-generation clinical candidate vector we have developed has the potential to achieve therapeutic effects in HoFH patients.

4.
Front Immunol ; 11: 1135, 2020.
Article in English | MEDLINE | ID: mdl-32733434

ABSTRACT

With the advent of single B-cell cloning technology, we can isolate antibodies against virtually any antigen to study the interaction of a given pathogen with the immune system and develop novel therapeutic strategies. Antibodies directed against the capsid of adeno-associated viruses (AAV) are a significant obstacle to effectively leveraging AAV as a gene-delivery vector in seropositive individuals. In order to design next-generation vectors that can evade neutralization by these antibodies, studies have mapped the epitopes of mouse monoclonal antibodies generated by immunization with AAV. Although these studies provide critical information regarding capsid immunogenicity, they cannot address (1) differences in the antibody repertoire generated in humans following AAV natural infection; or (2) how reactions can vary when generated in response to vector administration. Here, we isolated and evaluated a panel of novel, fully human anti-AAV antibodies by cloning single memory B cells from a seropositive normal donor. We have validated the utility of this approach to study AAV immunology. Our goal is to leverage this knowledge to design novel AAV variants that can effectively transduce target tissues in individuals with AAV-neutralizing antibodies.


Subject(s)
Antibodies, Monoclonal/isolation & purification , Antibodies, Viral/isolation & purification , B-Lymphocytes/immunology , Dependovirus/immunology , Immunologic Techniques , Clone Cells , Epitope Mapping , Genetic Therapy , Genetic Vectors/immunology , Humans , Immunologic Memory/immunology
5.
PLoS One ; 14(11): e0225188, 2019.
Article in English | MEDLINE | ID: mdl-31765413

ABSTRACT

Rare diseases defined by genetic mutations are classic targets for gene therapy. More recently, researchers expanded the use of gene therapy in non-clinical studies to infectious diseases through the delivery of vectorized antibodies to well-defined antigens. Here, we further extend the utility of gene therapy beyond the "accepted" indications to include organophosphate poisoning. There are no approved preventives for the multi-organ damage resulting from acute or chronic exposure to organophosphates. We show that a single intramuscular injection of adeno-associated virus vector produces peak expression (~0.5 mg/ml) of active human butyrylcholinesterase (hBChE) in mice serum within 3-4 weeks post-treatment. This expression is sustained for up to 140 days post-injection with no silencing. Sustained expression of hBChE provided dose-dependent protection against VX in male and female mice despite detectable antibodies to hBChE in some mice, thereby demonstrating that expression of hBChE in vivo in mouse muscle is an effective prophylactic against organophosphate poisoning.


Subject(s)
Butyrylcholinesterase/genetics , Dependovirus/genetics , Genetic Therapy/methods , Organophosphate Poisoning/therapy , Animals , Butyrylcholinesterase/metabolism , Female , Genetic Vectors/genetics , Humans , Male , Mice , Mice, Inbred C57BL
6.
Science ; 362(6414): 598-602, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30385580

ABSTRACT

Broadly neutralizing antibodies against highly variable pathogens have stimulated the design of vaccines and therapeutics. We report the use of diverse camelid single-domain antibodies to influenza virus hemagglutinin to generate multidomain antibodies with impressive breadth and potency. Multidomain antibody MD3606 protects mice against influenza A and B infection when administered intravenously or expressed locally from a recombinant adeno-associated virus vector. Crystal and single-particle electron microscopy structures of these antibodies with hemagglutinins from influenza A and B viruses reveal binding to highly conserved epitopes. Collectively, our findings demonstrate that multidomain antibodies targeting multiple epitopes exhibit enhanced virus cross-reactivity and potency. In combination with adeno-associated virus-mediated gene delivery, they may provide an effective strategy to prevent infection with influenza virus and other highly variable pathogens.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Camelids, New World/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A virus/immunology , Influenza B virus/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Animals , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/ultrastructure , Antibodies, Viral/chemistry , Antibodies, Viral/ultrastructure , Crystallography, X-Ray , Dogs , Female , Immunodominant Epitopes/chemistry , Immunodominant Epitopes/genetics , Immunodominant Epitopes/immunology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Neutralization Tests , Peptide Library , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Single-Domain Antibodies
7.
Cancer Res ; 78(21): 6171-6182, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30154145

ABSTRACT

Breast cancer brain metastases are a deadly sequela of primary breast tumors that overexpress human epidermal growth factor receptor 2 (HER2); median survival for patients with these tumors is 10 to 13 months from the time of diagnosis. Current treatments for HER2-positive breast cancer brain metastases are invasive, toxic, and largely ineffective. Here, we have developed an adeno-associated virus serotype 9 (AAV9) vector to express the anti-HER2 monoclonal antibody trastuzumab (Herceptin) in vivo A single prophylactic intrathecal administration of AAV9.trastuzumab vector in a novel orthotopic Rag1-/- murine xenograft model of HER2-positive breast cancer brain metastases significantly increased median survival, attenuated brain tumor growth, and preserved both the HER2 antigen specificity and the natural killer cell-associated mechanism of action of trastuzumab. When administered as a tumor treatment, AAV9.trastuzumab increased median survival. Dose-escalation studies revealed that higher doses of AAV9.trastuzumab resulted in smaller tumor volumes. Our results indicate that intrathecal AAV9.trastuzumab may provide significant antitumor activity in patients with HER2-positive breast cancer brain metastases.Significance: Intrathecal delivery of trastuzumab via adeno-associated virus has the potential to become a novel, integral part of adjuvant therapy for patients with HER2-positive breast cancer brain metastases. Cancer Res; 78(21); 6171-82. ©2018 AACR.


Subject(s)
Brain Neoplasms/therapy , Breast Neoplasms/therapy , Injections, Spinal/methods , Neoplasms/metabolism , Receptor, ErbB-2/metabolism , Trastuzumab/administration & dosage , Animals , Brain Neoplasms/secondary , Breast Neoplasms/pathology , Dependovirus/metabolism , Drug Delivery Systems , Female , Genetic Vectors , Homeodomain Proteins/genetics , Humans , Macaca , Macrophages/metabolism , Mice , Mice, Inbred NOD , Neoplasm Metastasis , Xenograft Model Antitumor Assays
8.
Hum Gene Ther Methods ; 29(3): 115-123, 2018 06.
Article in English | MEDLINE | ID: mdl-29848071

ABSTRACT

To evaluate gene therapy for retinal disorders, appropriate models of the human eye are needed. Nonhuman primate eyes offer significant advantages over rodent eyes. However, current preparation methods have limitations. Here, a protocol is described for histological processing of nonhuman primate eyes after gene transfer. The user dissects unfixed eyes, flattens the globe parts within filter paper, and performs formalin fixation and paraffin embedding. This method obviates the need for harsh fixatives, allowing subsequent immunostaining or in situ hybridization while preserving tissue integrity for histopathological evaluation. Moreover, the straight orientation of the retinal cell layers is ideal for image analysis.


Subject(s)
Gene Transfer Techniques , Genetic Therapy/methods , Histocytological Preparation Techniques/methods , Retina/metabolism , Animals , Histocytological Preparation Techniques/standards , Macaca fascicularis , Macaca mulatta
9.
Mol Ther ; 26(2): 542-549, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29292162

ABSTRACT

Sustained suppression of VEGF is needed in many patients with neovascular age-related macular degeneration (NVAMD), and gene transfer of a VEGF-neutralizing protein is a promising approach to achieve it. Initial clinical trials testing this approach have shown encouraging signals, but evidence of robust transgene expression and consistent antiangiogenic and antipermeability activity has been lacking. In this study, we demonstrate expression of an anti-human VEGF antibody fragment (antiVEGFfab) after subretinal injection of AAV8-antiVEGFfab. In transgenic mice expressing human VEGF in retina (rho/VEGF mice), a model of type 3 choroidal neovascularization (NV), eyes injected with ≥1 × 107 gene copies (GC) of AAV8-antiVEGFfab had significantly less mean area of NV than null vector-injected eyes. A dose-dependent response was observed with modest reduction of NV with ≤3 × 107, >50% reduction with ≥1 × 108 GC and almost complete elimination of NV with 3 × 109 or 1 × 1010 GC. In Tet/opsin/VEGF mice, in which doxycycline-induced high expression of VEGF leads to severe vascular leakage and exudative retinal detachment (RD), reduction of total RD by 70%-80% occurred with 3 × 109 or 1 × 1010 GC of AAV8-antiVEGFfab, an effect that was sustained for at least a month. These data strongly support initiating clinical trials testing subretinal injection of AAV8-antiVEGFfab in patients with NVAMD.


Subject(s)
Dependovirus/genetics , Genetic Vectors/genetics , Immunoglobulin Fab Fragments/genetics , Macular Degeneration/genetics , Macular Degeneration/pathology , Retinal Neovascularization/genetics , Retinal Neovascularization/pathology , Animals , Choroidal Neovascularization/genetics , Choroidal Neovascularization/pathology , Choroidal Neovascularization/therapy , Disease Models, Animal , Gene Expression , Gene Order , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors/administration & dosage , Immunoglobulin Fab Fragments/metabolism , Macular Degeneration/therapy , Mice , Retinal Neovascularization/therapy , Transduction, Genetic , Transgenes , Vascular Endothelial Growth Factor A/antagonists & inhibitors
10.
Vaccine ; 34(50): 6323-6329, 2016 12 07.
Article in English | MEDLINE | ID: mdl-27817961

ABSTRACT

The seroprevalence of neutralizing antibodies (NAbs) to adeno-associated viral (AAV) vector capsids may preclude a percentage of the population from receiving gene therapy, particularly following systemic vector administration. We hypothesized that the use of intramuscular (IM) administration of AAV vectors might circumvent this issue. IM injections were used to administer AAV8 vectors expressing either secreted or non-secreted transgenes into mice and the influence of NAbs supplied by pre-administration of pooled human IgG on transgene expression was evaluated. We then studied the impact of naturally occurring pre-existing AAV8 NAbs on expression of a secreted transgene following IM vector delivery in rhesus macaques. Finally, we evaluated the ability to readminister AAV vectors via IM injections in rhesus macaques. In mice, the presence of AAV8 NAbs had no effect on gene expression in the injected skeletal muscle. However, liver transgene expression following hepatic distribution of the vector was ablated. In rhesus macaques, naturally occurring pre-existing AAV8 NAb titers of ⩽1:160 had no effect on expression levels of a secreted transgene after IM delivery of the vector. Additionally, readministration of AAV vectors was possible by IM injection into the previously injected muscle groups, with no effect on transgene expression by the original vector. Therefore, the presence of pre-existing NAbs in the human population should not preclude subjects from receiving gene therapy by IM administration of the vector so long as sufficient levels of secreted transgene expression can be produced without the involvement of liver.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Dependovirus/immunology , Genetic Vectors/immunology , Animals , Gene Expression , Genetic Therapy/methods , Injections, Intramuscular , Macaca mulatta , Male , Mice, Inbred C57BL , Mice, Knockout , Seroepidemiologic Studies , Transgenes
11.
J Infect Dis ; 214(12): 1975-1979, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27683818

ABSTRACT

Adeno-associated viral vectors can be used as a platform for delivering biological countermeasures against pandemic and biological threats. We show that vector delivery of two antibody components of the ZMapp product is effective in mice against systemic and airway challenge with a mouse-adapted strain of Ebola virus. This platform provides a generic manufacturing solution and overcomes some of the delivery challenges associated with repeated administration of the protective protein.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Dependovirus/genetics , Drug Carriers , Gene Expression , Hemorrhagic Fever, Ebola/prevention & control , Immunologic Factors/biosynthesis , Animals , Antibodies, Monoclonal/genetics , Disease Models, Animal , Female , Genetic Therapy/methods , Hemorrhagic Fever, Ebola/immunology , Immunologic Factors/genetics , Mice, Inbred BALB C , Transduction, Genetic , Treatment Outcome
12.
PLoS One ; 9(11): e112268, 2014.
Article in English | MEDLINE | ID: mdl-25393537

ABSTRACT

Intramuscular (IM) administration of adeno-associated viral (AAV) vectors has entered the early stages of clinical development with some success, including the first approved gene therapy product in the West called Glybera. In preparation for broader clinical development of IM AAV vector gene therapy, we conducted detailed pre-clinical studies in mice and macaques evaluating aspects of delivery that could affect performance. We found that following IM administration of AAV8 vectors in mice, a portion of the vector reached the liver and hepatic gene expression contributed significantly to total expression of secreted transgenes. The contribution from liver could be controlled by altering injection volume and by the use of traditional (promoter) and non-traditional (tissue-specific microRNA target sites) expression control elements. Hepatic distribution of vector following IM injection was also noted in rhesus macaques. These pre-clinical data on AAV delivery should inform safe and efficient development of future AAV products.


Subject(s)
Dependovirus/genetics , Genetic Vectors/administration & dosage , Liver/metabolism , Transgenes , Animals , Gene Expression , Gene Transfer Techniques , Injections, Intramuscular , Macaca mulatta , Male , Mice , MicroRNAs/metabolism , Organ Specificity
13.
Clin Vaccine Immunol ; 21(11): 1528-33, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25209558

ABSTRACT

Influenza causes serious and sometimes fatal disease in individuals at risk due to advanced age or immunodeficiencies. Despite progress in the development of seasonal influenza vaccines, vaccine efficacy in elderly and immunocompromised individuals remains low. We recently developed a passive immunization strategy using an adeno-associated virus (AAV) vector to deliver a neutralizing anti-influenza antibody at the site of infection, the nasal airways. Here we show that young, old, and immunodeficient (severe combined immunodeficient [SCID]) mice that were treated intranasally with AAV9 vector expressing a modified version of the broadly neutralizing anti-influenza antibody FI6 were protected and exhibited no signs of disease following an intranasal challenge with the mouse-adapted H1N1 influenza strain A/Puerto Rico/8/1934(H1N1) (PR8) (Mt. Sinai strain). Nonvaccinated mice succumbed to the PR8 challenge due to severe weight loss. We propose that airway-directed AAV9 passive immunization against airborne infectious agents may be beneficial in elderly and immunocompromised patients, for whom there still exists an unmet need for effective vaccination against influenza.


Subject(s)
Antibodies, Viral/immunology , Biological Therapy/methods , Dependovirus/growth & development , Drug Carriers/administration & dosage , Immunization, Passive/methods , Influenza A Virus, H1N1 Subtype/immunology , Orthomyxoviridae Infections/prevention & control , Administration, Intranasal , Animals , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , Antibodies, Viral/genetics , Dependovirus/genetics , Disease Models, Animal , Female , Immunocompromised Host , Mice, Inbred BALB C , Mice, SCID , Survival Analysis , Treatment Outcome
14.
Proc Natl Acad Sci U S A ; 110(41): 16538-43, 2013 Oct 08.
Article in English | MEDLINE | ID: mdl-24043801

ABSTRACT

Effective control of HIV-1 infection in humans is achieved using combinations of antiretroviral therapy (ART) drugs. In humanized mice (hu-mice), control of viremia can be achieved using either ART or by immunotherapy using combinations of broadly neutralizing antibodies (bNAbs). Here we show that treatment of HIV-1-infected hu-mice with a combination of three highly potent bNAbs not only resulted in complete viremic control but also led to a reduction in cell-associated HIV-1 DNA. Moreover, lowering the initial viral load by coadministration of ART and immunotherapy enabled prolonged viremic control by a single bNAb after ART was withdrawn. Similarly, a single injection of adeno-associated virus directing expression of one bNAb produced durable viremic control after ART was terminated. We conclude that immunotherapy reduces plasma viral load and cell-associated HIV-1 DNA and that decreasing the initial viral load enables single bNAbs to control viremia in hu-mice.


Subject(s)
Anti-Retroviral Agents/immunology , Antibodies, Neutralizing/immunology , HIV Infections/prevention & control , HIV-1/drug effects , HIV-1/immunology , Immunotherapy/methods , Animals , Anti-Retroviral Agents/pharmacology , Antibodies, Neutralizing/pharmacology , DNA Primers/genetics , DNA, Viral/metabolism , Dependovirus , Drug Therapy, Combination , Humans , Mice , Mice, Transgenic , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA , Viral Load/drug effects
15.
Sci Transl Med ; 5(187): 187ra72, 2013 May 29.
Article in English | MEDLINE | ID: mdl-23720583

ABSTRACT

The emergence of a new influenza pandemic remains a threat that could result in a substantial loss of life and economic disruption worldwide. Advances in human antibody isolation have led to the discovery of monoclonal antibodies (mAbs) that have broad neutralizing activity against various influenza strains, although their direct use for prophylaxis is impractical. To overcome this limitation, our approach is to deliver antibody via adeno-associated virus (AAV) vectors to the site of initial infection, which, for respiratory viruses such as influenza, is the nasopharyngeal mucosa. AAV vectors based on serotype 9 were engineered to express a modified version of the previously isolated broadly neutralizing mAb to influenza A, FI6. We demonstrate that intranasal delivery of AAV9.FI6 into mice afforded complete protection and log reductions in viral load to 100 LD50 (median lethal dose) of three clinical isolates of H5N1 and two clinical isolates of H1N1, all of which have been associated with historic human pandemics (including H1N1 1918). Similarly, complete protection was achieved in ferrets challenged with lethal doses of H5N1 and H1N1. This approach serves as a platform for the prevention of natural or deliberate respiratory diseases for which a protective antibody is available.


Subject(s)
Antibodies, Neutralizing/genetics , Antibodies, Viral/genetics , Gene Transfer Techniques , Orthomyxoviridae Infections/prevention & control , Administration, Intranasal , Animals , Antibodies, Neutralizing/administration & dosage , Antibodies, Viral/administration & dosage , Dependovirus/genetics , Dose-Response Relationship, Immunologic , Female , Ferrets , Macaca mulatta , Mice , Mice, Inbred BALB C
16.
J Neuroimmunol ; 181(1-2): 68-81, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17011639

ABSTRACT

Serotonin plays a role in T cell activation, but there is no clear consensus of which of the 14 serotonergic receptors control this activations pathway. We have used a broad range of serotonergic receptor antagonists to define the functional involvement of these receptors governing the proliferation of primary T cells as well as in T cell lines. Our data shows that antagonism of the 5-HT(1B) receptor inhibits the proliferation of both human and murine primary helper T cells and of human helper T cell lines. As a whole, our data suggest that other serotonergic receptors may contribute to the proliferative signals, but the 5-HT(1B) receptor plays the most dominant role.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Receptor, Serotonin, 5-HT1B/metabolism , Animals , Apoptosis/drug effects , Apoptosis/immunology , Cell Division/immunology , Chlorocebus aethiops , Flow Cytometry , Gene Expression/immunology , Humans , Jurkat Cells , Lymphocyte Activation/physiology , Muridae , Receptor, Serotonin, 5-HT1B/genetics , Serotonin/metabolism , Serotonin 5-HT1 Receptor Antagonists , Serotonin Antagonists/pharmacology
18.
Nucleic Acids Res ; 31(14): 4218-26, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12853640

ABSTRACT

The yeast protein Rrf1p encoded by the FIL1 nuclear gene bears significant sequence similarity to Escherichia coli ribosome recycling factor (RRF). Here, we call FIL1 Ribosome Recycling Factor of yeast, RRF1. Its gene product, Rrf1p, was localized in mitochondria. Deletion of RRF1 leads to a respiratory incompetent phenotype and to instability of the mitochondrial genome (conversion to rho(-)/rho(0) cytoplasmic petites). Yeast with intact mitochondria and with deleted genomic RRF1 that harbors a plasmid carrying RRF1 was prepared from spores of heterozygous diploid yeast. Such yeast with a mutated allele of RRF1, rrf1-L209P, grew on a non-fermentable carbon source at 30 but not at 36 degrees C, where mitochondrial but not total protein synthesis was 90% inhibited. We propose that Rrf1p is essential for mitochondrial protein synthesis and acts as a RRF in mitochondria.


Subject(s)
Mitochondrial Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Cell Division/genetics , DNA, Mitochondrial/genetics , Electron Transport/genetics , Gene Deletion , Genetic Complementation Test , Mitochondria/genetics , Mitochondria/physiology , Mutation , Saccharomyces cerevisiae/growth & development , Temperature
19.
Acta Crystallogr D Biol Crystallogr ; 59(Pt 6): 1114-6, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12777792

ABSTRACT

Detergents are required to solubilize integral membrane proteins and are common components of the solutions used to crystallize these molecules. It has been unclear whether these detergents are completely compatible with the oils used in the microbatch-under-oil crystallization technique, because they might conceivably be lost from solution by partitioning into the oil phase. The partitioning of the detergents n-octyl-beta-d-glucopyranoside and Fos-Choline-12 into two different oils used for microbatch crystallization experiments has been examined. It was found that vigorous mixing and prolonged incubation of the aqueous detergent solutions with the oils leads to small losses of detergent (approximately 5% of the total detergent mass); however, gentle mixing that is more typical of the mixing encountered in a crystallization experiments leads to negligible loss of detergent.


Subject(s)
Crystallization/methods , Detergents/chemistry , Glucosides/chemistry , Oils/chemistry , Glycerylphosphorylcholine/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...