Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Bioanalysis ; 5(9): 1041-55, 2013 May.
Article in English | MEDLINE | ID: mdl-23641695

ABSTRACT

BACKGROUND: Immunogenicity testing is an important component of clinical development for large-molecule biotherapeutics. New complex types of large molecules, such as antibody-drug conjugates (ADCs), require careful evaluation of the testing strategy and bioanalytical assays used to monitor the development of antitherapeutic antibodies. RESULTS: An electrochemiluminescence-based immunoassay for the detection and epitope characterization of anti-ADC antibodies was validated. Using this assay format, antibodies directed against the monoclonal antibody and linker-drug components of the ADC were successfully detected in a multiple-dose rat toxicity study. CONCLUSION: Immunogenicity assays incorporating epitope determination may provide additional information about the characteristics of induced antitherapeutic antibodies, including the magnitude and timing of the various types of antibody responses.


Subject(s)
Antibodies, Monoclonal/blood , Antibodies, Monoclonal/immunology , Immunoconjugates/blood , Immunoconjugates/immunology , Immunogenetic Phenomena/immunology , Pharmaceutical Preparations/blood , Animals , Antibodies, Monoclonal/therapeutic use , Antigen-Antibody Reactions , Humans , Immunoconjugates/therapeutic use
2.
AAPS J ; 15(1): 30-40, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23054969

ABSTRACT

The availability of highly sensitive immunoassays enables the detection of antidrug antibody (ADA) responses of various concentrations and affinities. The analysis of the impact of antibody status on drug pharmacokinetics (PK) is confounded by the presence of low-affinity or low-concentration antibody responses within the dataset. In a phase 2 clinical trial, a large proportion of subjects (45%) developed ADA following weekly dosing with AMG 317, a fully human monoclonal antibody therapeutic. The antibody responses displayed a wide range of relative concentrations (30 ng/mL to >13 µg/mL) and peaked at various times during the study. To evaluate the impact of immunogenicity on PK, AMG 317 concentration data were analyzed following stratification by dose group, time point, antibody status (positive or negative), and antibody level (relative concentration). With dose group as a stratifying variable, a moderate reduction in AMG 317 levels (<50%) was observed in antibody-positive subjects when compared to antibody-negative subjects, but the difference was not statistically significant in all dose groups. The most significant reduction in AMG 317 levels was revealed when antibody data was stratified by both time point and antibody level. In general, high ADA concentrations (>500 ng/mL) and later time points (week 12) were associated with significantly (up to 97%) lower trough AMG 317 concentrations. The use of quasi-quantitative antibody data and appropriate statistical methods was critical for the most comprehensive evaluation of the impact of immunogenicity on PK.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies/blood , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized , Humans , Patient Satisfaction
3.
BMC Clin Pharmacol ; 11: 17, 2011 Nov 09.
Article in English | MEDLINE | ID: mdl-22070868

ABSTRACT

BACKGROUND: Panitumumab is a fully human antibody against the epidermal growth factor receptor that is indicated for the treatment of metastatic colorectal cancer (mCRC) after disease progression on standard chemotherapy. The purpose of this analysis was to examine the immunogenicity of panitumumab and to evaluate the effect of anti-panitumumab antibodies on pharmacokinetic and safety profiles in patients with mCRC receiving panitumumab in combination with oxaliplatin- or irinotecan-based chemotherapies. METHODS: Three validated assays (two screening immunoassays and a neutralizing antibody bioassay) were used to detect the presence of anti-panitumumab antibodies in serum samples collected from patients enrolled in four panitumumab combination chemotherapy clinical trials. The impact of anti-panitumumab antibodies on pharmacokinetic and safety profiles was analyzed using population pharmacokinetic analysis and descriptive statistics, respectively. RESULTS: Of 1124 patients treated with panitumumab in combination with oxaliplatin- or irinotecan-based chemotherapy with postbaseline samples available for testing, 20 (1.8%) patients developed binding antibodies and 2 (0.2%) developed neutralizing antibodies. The incidence of anti-panitumumab antibodies was similar in patients with tumors expressing wild-type or mutant KRAS and in patients receiving oxaliplatin- or irinotecan-based chemotherapies. No evidence of an altered pharmacokinetic or safety profile was found in patients who tested positive for anti-panitumumab antibodies. CONCLUSIONS: The immunogenicity of panitumumab in the combination chemotherapy setting was infrequent and similar to the immunogenicity observed in the monotherapy setting. Panitumumab immunogenicity did not appear to alter pharmacokinetic or safety profiles. This low rate of immunogenicity may be attributed to the fully human nature of panitumumab.


Subject(s)
Antibodies, Anti-Idiotypic/analysis , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal/adverse effects , Antineoplastic Agents/adverse effects , Drug Hypersensitivity/immunology , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/blood , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Neutralizing/analysis , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Colorectal Neoplasms/complications , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/immunology , Drug Hypersensitivity/blood , Drug Hypersensitivity/complications , Drug Hypersensitivity/epidemiology , Humans , Incidence , Irinotecan , Organoplatinum Compounds/therapeutic use , Oxaliplatin , Panitumumab , Severity of Illness Index
4.
Pharm Res ; 28(10): 2530-42, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21604075

ABSTRACT

PURPOSE: For AMG 317, a fully human monoclonal antibody to interleukin receptor IL-4Rα, we developed a population pharmacokinetic (PK) model by fitting data from four early phase clinical trials of intravenous and subcutaneous (SC) routes simultaneously, investigated important PK covariates, and explored the relationship between exposure and IgE response. METHODS: Data for 294 subjects and 2183 AMG 317 plasma concentrations from three Phase 1 and 1 Phase 2 studies were analyzed by nonlinear mixed effects modeling using first-order conditional estimation with interaction. The relationship of IgE response with post hoc estimates of exposure generated from the final PK model was explored based on data from asthmatic patients. RESULTS: The best structural model was a two-compartment quasi-steady-state target-mediated drug disposition model with linear and non-linear clearances. For a typical 80-kg, 40-year subject, linear clearance was 35.0 mL/hr, central and peripheral volumes of distribution were 1.78 and 5.03 L, respectively, and SC bioavailability was 24.3%. Body weight was an important covariate on linear clearance and central volume; age influenced absorption rate. A significant treatment effect was observable between the cumulative AUC and IgE response measured. CONCLUSION: The population PK model adequately described AMG 317 PK from IV and SC routes over a 60-fold range of doses with two dosing strengths across multiple studies covering healthy volunteers and patients with mild to severe asthma. IgE response across a range of doses and over the sampling time points was found to be related to cumulative AMG 317 exposure.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/pharmacokinetics , Immunoglobulin E/metabolism , Interleukin-4 Receptor alpha Subunit/immunology , Adult , Antibodies, Monoclonal/blood , Antibodies, Monoclonal, Humanized , Area Under Curve , Asthma/drug therapy , Asthma/metabolism , Biological Availability , Body Weight , Dose-Response Relationship, Drug , Female , Humans , Infusions, Intravenous , Infusions, Subcutaneous , Interleukin-4 Receptor alpha Subunit/antagonists & inhibitors , Linear Models , Male , Models, Biological , Nonlinear Dynamics
5.
Hum Mol Genet ; 19(5): 861-78, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20015953

ABSTRACT

Biogenesis of lysosome-related organelles complex 1 (BLOC-1) is a protein complex formed by the products of eight distinct genes. Loss-of-function mutations in two of these genes, DTNBP1 and BLOC1S3, cause Hermansky-Pudlak syndrome, a human disorder characterized by defective biogenesis of lysosome-related organelles. In addition, haplotype variants within the same two genes have been postulated to increase the risk of developing schizophrenia. However, the molecular function of BLOC-1 remains unknown. Here, we have generated a fly model of BLOC-1 deficiency. Mutant flies lacking the conserved Blos1 subunit displayed eye pigmentation defects due to abnormal pigment granules, which are lysosome-related organelles, as well as abnormal glutamatergic transmission and behavior. Epistatic analyses revealed that BLOC-1 function in pigment granule biogenesis requires the activities of BLOC-2 and a putative Rab guanine-nucleotide-exchange factor named Claret. The eye pigmentation phenotype was modified by misexpression of proteins involved in intracellular protein trafficking; in particular, the phenotype was partially ameliorated by Rab11 and strongly enhanced by the clathrin-disassembly factor, Auxilin. These observations validate Drosophila melanogaster as a powerful model for the study of BLOC-1 function and its interactions with modifier genes.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Eye Proteins/genetics , Animals , Animals, Genetically Modified , Drosophila melanogaster/metabolism , Hermanski-Pudlak Syndrome/genetics , Hermanski-Pudlak Syndrome/metabolism , Humans , Models, Animal , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Organelles/metabolism , Phenotype
6.
Mol Genet Metab ; 93(2): 134-44, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17933573

ABSTRACT

Hermansky-Pudlak syndrome (HPS) comprises a constellation of human autosomal recessive disorders characterized by albinism and platelet storage pool deficiency. At least eight types of HPS have been defined based on the identity of the mutated gene. These genes encode components of four ubiquitously expressed protein complexes, named Adaptor Protein (AP)-3 and Biogenesis of Lysosome-related Organelles Complex (BLOC)-1 through -3. In patients of Puerto Rican origin, the molecular diagnosis can be based on analysis of two founder mutations. On the other hand, identification of the HPS type in other patients relies on the sequencing of all candidate genes. In this work, we have developed a biochemical assay to minimize the number of candidate genes to be sequenced per patient. The assay consists of immunoblotting analysis of extracts prepared from skin fibroblasts, using antibodies to one subunit per protein complex. The assay allowed us to determine which complex was defective in each of a group of HPS patients with unknown genetic lesions, thus subsequent sequencing was limited to genes encoding the corresponding subunits. Because no mutations within the two genes encoding BLOC-3 subunits could be found in two patients displaying reduced BLOC-3 levels, the possible existence of additional subunits was considered. Through size-exclusion chromatography and sedimentation velocity analysis, the native molecular mass of BLOC-3 was estimated to be 140+/-30 kDa, a value most consistent with the idea that BLOC-3 is a HPS1HPS4 heterodimer (approximately 156 kDa) albeit not inconsistent with the putative existence of a relatively small third subunit.


Subject(s)
Blotting, Western/methods , Hermanski-Pudlak Syndrome/diagnosis , Hermanski-Pudlak Syndrome/genetics , Adaptor Protein Complex 3/genetics , Adaptor Protein Complex 3/immunology , Amino Acid Sequence , Animals , Antibody Specificity , Base Sequence , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/immunology , DNA/genetics , Hermanski-Pudlak Syndrome/classification , Humans , Mice , Molecular Sequence Data , Molecular Weight , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/immunology , Proteins/genetics , Proteins/immunology , Sequence Homology, Amino Acid
7.
Mol Biol Cell ; 18(3): 768-80, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17182842

ABSTRACT

Hermansky-Pudlak syndrome (HPS) is a genetic disorder characterized by defects in the formation and function of lysosome-related organelles such as melanosomes. HPS in humans or mice is caused by mutations in any of 15 genes, five of which encode subunits of biogenesis of lysosome-related organelles complex (BLOC)-1, a protein complex with no known function. Here, we show that BLOC-1 functions in selective cargo exit from early endosomes toward melanosomes. BLOC-1-deficient melanocytes accumulate the melanosomal protein tyrosinase-related protein-1 (Tyrp1), but not other melanosomal proteins, in endosomal vacuoles and the cell surface due to failed biosynthetic transit from early endosomes to melanosomes and consequent increased endocytic flux. The defects are corrected by restoration of the missing BLOC-1 subunit. Melanocytes from HPS model mice lacking a different protein complex, BLOC-2, accumulate Tyrp1 in distinct downstream endosomal intermediates, suggesting that BLOC-1 and BLOC-2 act sequentially in the same pathway. By contrast, intracellular Tyrp1 is correctly targeted to melanosomes in melanocytes lacking another HPS-associated protein complex, adaptor protein (AP)-3. The results indicate that melanosome maturation requires at least two cargo transport pathways directly from early endosomes to melanosomes, one pathway mediated by AP-3 and one pathway mediated by BLOC-1 and BLOC-2, that are deficient in several forms of HPS.


Subject(s)
Endosomes/metabolism , Lysosomes/metabolism , Multiprotein Complexes/metabolism , Vacuoles/metabolism , Adaptor Protein Complex 3/metabolism , Amino Acid Sequence , Animals , Endocytosis , Endosomes/ultrastructure , Humans , Lysosomes/ultrastructure , Melanins/biosynthesis , Melanosomes/ultrastructure , Membrane Glycoproteins/chemistry , Mice , Mice, Inbred C57BL , Models, Biological , Molecular Sequence Data , Monophenol Monooxygenase/metabolism , Mutant Proteins/metabolism , Oxidoreductases/chemistry , Pigmentation/physiology , Protein Transport , Qa-SNARE Proteins/metabolism , Vacuoles/ultrastructure
8.
Biochem J ; 395(3): 587-98, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16448387

ABSTRACT

Dysbindin was identified as a dystrobrevin-binding protein potentially involved in the pathogenesis of muscular dystrophy. Subsequently, genetic studies have implicated variants of the human dysbindin-encoding gene, DTNBP1, in the pathogeneses of Hermansky-Pudlak syndrome and schizophrenia. The protein is a stable component of a multisubunit complex termed BLOC-1 (biogenesis of lysosome-related organelles complex-1). In the present study, the significance of the dystrobrevin-dysbindin interaction for BLOC-1 function was examined. Yeast two-hybrid analyses, and binding assays using recombinant proteins, demonstrated direct interaction involving coiled-coil-forming regions in both dysbindin and the dystrobrevins. However, recombinant proteins bearing the coiled-coil-forming regions of the dystrobrevins failed to bind endogenous BLOC-1 from HeLa cells or mouse brain or muscle, under conditions in which they bound the Dp71 isoform of dystrophin. Immunoprecipitation of endogenous dysbindin from brain or muscle resulted in robust co-immunoprecipitation of the pallidin subunit of BLOC-1 but no specific co-immunoprecipitation of dystrobrevin isoforms. Within BLOC-1, dysbindin is engaged in interactions with three other subunits, named pallidin, snapin and muted. We herein provide evidence that the same 69-residue region of dysbindin that is sufficient for dystrobrevin binding in vitro also contains the binding sites for pallidin and snapin, and at least part of the muted-binding interface. Functional, histological and immunohistochemical analyses failed to detect any sign of muscle pathology in BLOC-1-deficient, homozygous pallid mice. Taken together, these results suggest that dysbindin assembled into BLOC-1 is not a physiological binding partner of the dystrobrevins, likely due to engagement of its dystrobrevin-binding region in interactions with other subunits.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Dystrophin-Associated Proteins/metabolism , Animals , Carrier Proteins/genetics , Dystrophin/deficiency , Dystrophin/genetics , Dystrophin/metabolism , HeLa Cells , Humans , Immunohistochemistry , Lectins/metabolism , Mice , Mice, Knockout , Molecular Sequence Data , Muscles/cytology , Muscles/metabolism , Nerve Tissue Proteins , Protein Binding , Protein Subunits/genetics , Protein Subunits/metabolism , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Two-Hybrid System Techniques
9.
Traffic ; 6(3): 243-51, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15702992

ABSTRACT

Melanocytes and cells of the immune system share an unusual secretory mechanism which uses the lysosome as a regulated secretory organelle. Recently, a number of the proteins required for these 'secretory lysosomes' to undergo exocytosis have been identified. These include Rab27a, Lyst, Rab geranyl geranyl transferase and the adapter protein complex AP-3. Patients lacking any of these proteins are characterized by the rare combination of albinism and immunodeficiency, revealing roles for these proteins in both melanocyte and immune cell secretion. In order to ask how far the link between albinism and immunodeficiency extends we have examined cytotoxic T-lymphocyte (CTL) secretion from two BLOC-3-deficient patients and seven different mouse models of Hermansky-Pudlak syndrome, all of which display defects in pigmentation and platelet function. We find that CTL function is normal in HPS patients and pale-ear mice deficient in BLOC-3, pallid, muted and sandy mice deficient in BLOC-1, ruby-eye mice deficient in BLOC-2 and buff mice deficient in Vps33a. Similarly, the unconventional myosins, Va, VIIa and XV, which can act as effectors for Rab27a in some cell types, are not required in CTL. These results reveal differences in the protein machinery required for biogenesis and/or secretion of lysosome-related organelles in CTL and melanocytes.


Subject(s)
Carrier Proteins/physiology , Hermanski-Pudlak Syndrome/physiopathology , Lysosomes/metabolism , Myosins/deficiency , T-Lymphocytes, Cytotoxic/metabolism , Animals , Dyneins/deficiency , Dyneins/physiology , Humans , Melanocytes/metabolism , Mice , Mice, Mutant Strains , Molecular Motor Proteins , Myosin Heavy Chains/deficiency , Myosin Heavy Chains/physiology , Myosin Type V/deficiency , Myosin Type V/physiology , Myosin VIIa , Myosins/physiology , Secretory Vesicles/physiology
10.
Environ Health Perspect ; 112(6): 672-7, 2004 May.
Article in English | MEDLINE | ID: mdl-15121509

ABSTRACT

Some organochlorine pesticides and other synthetic chemicals mimic hormones in representatives of each vertebrate class, including mammals, reptiles, amphibians, birds, and fish. These compounds are called endocrine-disrupting chemicals (EDCs). Similarly, hormonelike signaling has also been observed when vertebrates are exposed to plant chemicals called phytoestrogens. Previous research has shown the mechanism of action for EDCs and phytoestrogens is as unintended ligands for the estrogen receptor (ER). Although pesticides have been synthesized to deter insects and weeds, plants produce phytoestrogens to deter herbivores, as attractant cues for insects, and as recruitment signals for symbiotic soil bacteria. Our data present the first evidence that some of the same organochlorine pesticides and EDCs known to disrupt endocrine signaling through ERs in exposed wildlife and humans also disrupt the phytoestrogen signaling that leguminous plants use to recruit Sinorhizobium meliloti soil bacteria for symbiotic nitrogen fixation. Here we report that a variety of EDCs and pesticides commonly found in agricultural soils interfere with the symbiotic signaling necessary for nitrogen fixation, suggesting that the principles underlying endocrine disruption may have more widespread biological and ecological importance than had once been thought.


Subject(s)
Endocrine System/drug effects , Environmental Pollutants/pharmacology , Environmental Pollutants/poisoning , Fabaceae/microbiology , Fabaceae/physiology , Gene Expression Regulation/drug effects , Hydrocarbons, Chlorinated , Insecticides/poisoning , Isoflavones/pharmacology , Plant Preparations/pharmacology , Rhizobium/physiology , Signal Transduction , Symbiosis , Cell Communication/drug effects , Fabaceae/genetics , Phytoestrogens , Receptors, Estrogen/drug effects , Transcriptional Activation
11.
J Biol Chem ; 279(27): 28393-401, 2004 Jul 02.
Article in English | MEDLINE | ID: mdl-15102850

ABSTRACT

Biogenesis of lysosome-related organelles complex-1 (BLOC-1) is a ubiquitously expressed multisubunit protein complex required for the normal biogenesis of specialized organelles of the endosomal-lysosomal system, such as melanosomes and platelet dense granules. The complex is known to contain the coiled-coil-forming proteins, Pallidin, Muted, Cappuccino, and Dysbindin. The genes encoding these proteins are defective in inbred mouse strains that serve as models of Hermansky-Pudlak syndrome (HPS), a genetic disorder characterized by hypopigmentation and platelet storage pool deficiency. In addition, mutation of human Dysbindin causes HPS type 7. Here, we report the identification of another four subunits of the complex. One is Snapin, a coiled-coil-forming protein previously characterized as a binding partner of synaptosomal-associated proteins 25 and 23 and implicated in the regulation of membrane fusion events. The other three are previously uncharacterized proteins, which we named BLOC subunits 1, 2, and 3 (BLOS1, -2, and -3). Using specific antibodies to detect endogenous proteins from human and mouse cells, we found that Snapin, BLOS1, BLOS2, and BLOS3 co-immunoprecipitate, and co-fractionate upon size exclusion chromatography, with previously known BLOC-1 subunits. Furthermore, steady-state levels of the four proteins are significantly reduced in cells from pallid mice, which carry a mutation in Pallidin and display secondary loss of other BLOC-1 subunits. Yeast two-hybrid analyses suggest a network of binary interactions involving all of the previously known and newly identified subunits. Interestingly, the HPS mouse model strain, reduced pigmentation, carries a nonsense mutation in the gene encoding BLOS3. As judged from size exclusion chromatographic analyses, the reduced pigmentation mutation affects BLOC-1 assembly less severely than the pallid mutation. Mutations in the human genes encoding Snapin and the BLOS proteins could underlie novel forms of HPS.


Subject(s)
Carrier Proteins/biosynthesis , Carrier Proteins/physiology , Membrane Proteins/biosynthesis , Vesicular Transport Proteins , Animals , Blood Platelets/metabolism , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cattle , Chromatography , Codon, Nonsense , Cytosol/metabolism , DNA Mutational Analysis , Dysbindin , Dystrophin-Associated Proteins , Electrophoresis, Polyacrylamide Gel , Gene Expression Regulation , HeLa Cells , Hermanski-Pudlak Syndrome/genetics , Humans , Immunoblotting , Immunohistochemistry , Intracellular Signaling Peptides and Proteins , Lectins , Liver/metabolism , Mass Spectrometry , Melanosomes/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Models, Biological , Molecular Sequence Data , Mutation , Nerve Tissue Proteins/metabolism , Phenotype , Precipitin Tests , Protein Binding , Proteins , Qb-SNARE Proteins , Qc-SNARE Proteins , RNA, Messenger/metabolism , Recombinant Fusion Proteins/metabolism , Synaptosomal-Associated Protein 25 , Two-Hybrid System Techniques
12.
Nat Genet ; 35(1): 84-9, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12923531

ABSTRACT

Hermansky-Pudlak syndrome (HPS; MIM 203300) is a genetically heterogeneous disorder characterized by oculocutaneous albinism, prolonged bleeding and pulmonary fibrosis due to abnormal vesicle trafficking to lysosomes and related organelles, such as melanosomes and platelet dense granules. In mice, at least 16 loci are associated with HPS, including sandy (sdy; ref. 7). Here we show that the sdy mutant mouse expresses no dysbindin protein owing to a deletion in the gene Dtnbp1 (encoding dysbindin) and that mutation of the human ortholog DTNBP1 causes a novel form of HPS called HPS-7. Dysbindin is a ubiquitously expressed protein that binds to alpha- and beta-dystrobrevins, components of the dystrophin-associated protein complex (DPC) in both muscle and nonmuscle cells. We also show that dysbindin is a component of the biogenesis of lysosome-related organelles complex 1 (BLOC-1; refs. 9-11), which regulates trafficking to lysosome-related organelles and includes the proteins pallidin, muted and cappuccino, which are associated with HPS in mice. These findings show that BLOC-1 is important in producing the HPS phenotype in humans, indicate that dysbindin has a role in the biogenesis of lysosome-related organelles and identify unexpected interactions between components of DPC and BLOC-1.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/genetics , Dystrophin-Associated Proteins , Hermanski-Pudlak Syndrome/genetics , Mutation , Animals , COS Cells , Carrier Proteins/metabolism , Cytoskeletal Proteins/metabolism , Dysbindin , Female , Humans , Intracellular Signaling Peptides and Proteins , Lectins , Macromolecular Substances , Male , Melanosomes/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Middle Aged , Molecular Sequence Data , Phosphoproteins/metabolism , Protein Binding
13.
Semin Cell Dev Biol ; 13(4): 271-8, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12243726

ABSTRACT

Hermansky-Pudlak syndrome (HPS) defines a group of autosomal recessive disorders characterized by defects in lysosome-related organelles such as melanosomes and platelet dense granules. The genes that are defective in each of the different forms of HPS in humans, or in HPS-like disorders in mice, are thought to encode components of a putative molecular machinery required for the formation of specialized organelles of the lysosomal system. This review discusses the biochemical and functional properties of the products of identified HPS genes, which include subunits of the AP-3 complex and the novel proteins HPS1p, HPS3p, HPS4p, pallidin and muted.


Subject(s)
Hermanski-Pudlak Syndrome/metabolism , Hermanski-Pudlak Syndrome/pathology , Lysosomes/metabolism , Lysosomes/pathology , Humans , Melanosomes/metabolism , Melanosomes/pathology , Protein Transport/physiology
14.
J Biol Chem ; 277(31): 28191-9, 2002 Aug 02.
Article in English | MEDLINE | ID: mdl-12019270

ABSTRACT

Recent studies have led to the identification of a group of genes required for normal biogenesis of lysosome-related organelles such as melanosomes and platelet-dense granules. Two of these genes, which are defective in the pallid and muted mutant mouse strains, encode small, coiled-coil-forming proteins that display no homology to each other or to any known protein. We report that these two proteins, pallidin and muted, are components of a novel protein complex. We raised antibodies that allow for detection of pallidin from a wide variety of mammalian cells. Endogenous pallidin was distributed in both soluble and peripheral membrane protein fractions. Size-exclusion chromatography and sedimentation velocity analyses indicated that the bulk of cytosolic pallidin is a component of an asymmetric protein complex with a molecular mass of approximately 200 kDa. We named this complex BLOC-1 (for biogenesis of lysosome-related organelles complex 1). Steady-state pallidin protein levels were reduced in fibroblasts derived from muted and reduced pigmentation mice, suggesting that the genes defective in these two mutant strains could encode components of BLOC-1 that are required for pallidin stability. Co-immunoprecipitation and immunodepletion experiments using an antibody to muted confirmed that this protein is a subunit of BLOC-1. Yeast two-hybrid analyses revealed that pallidin is capable of self-association through a region that contains its two coiled-coil forming domains. Unlike AP-3-deficient pearl fibroblasts, which display defects in intracellular zinc storage, zinc distribution was not noticeably affected in pallid or muted fibroblasts. Interestingly, immunofluorescence and in vitro binding experiments demonstrated that pallidin/BLOC-1 is able to associate with actin filaments. We propose that BLOC-1 mediates the biogenesis of lysosome-related organelles by a mechanism that may involve self-assembly and interaction with the actin cytoskeleton.


Subject(s)
Blood Platelets/physiology , Carrier Proteins/metabolism , Cytoplasmic Granules/physiology , Melanosomes/physiology , Proteins/metabolism , Actins/metabolism , Animals , Carrier Proteins/chemistry , Cells, Cultured , Fibroblasts/cytology , Humans , Intracellular Signaling Peptides and Proteins , Lectins , Macromolecular Substances , Mice , Mice, Mutant Strains , Microscopy, Fluorescence , Skin/cytology , Zinc/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...