Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Proc Natl Acad Sci U S A ; 115(31): E7379-E7388, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30018062

ABSTRACT

The precise correction of genetic mutations at the nucleotide level is an attractive permanent therapeutic strategy for human disease. However, despite significant progress, challenges to efficient and accurate genome editing persist. Here, we report a genome editing platform based upon a class of hematopoietic stem cell (HSC)-derived clade F adeno-associated virus (AAV), which does not require prior nuclease-mediated DNA breaks and functions exclusively through BRCA2-dependent homologous recombination. Genome editing is guided by complementary homology arms and is highly accurate and seamless, with no evidence of on-target mutations, including insertion/deletions or inclusion of AAV inverted terminal repeats. Efficient genome editing was demonstrated at different loci within the human genome, including a safe harbor locus, AAVS1, and the therapeutically relevant IL2RG gene, and at the murine Rosa26 locus. HSC-derived AAV vector (AAVHSC)-mediated genome editing was robust in primary human cells, including CD34+ cells, adult liver, hepatic endothelial cells, and myocytes. Importantly, high-efficiency gene editing was achieved in vivo upon a single i.v. injection of AAVHSC editing vectors in mice. Thus, clade F AAV-mediated genome editing represents a promising, highly efficient, precise, single-component approach that enables the development of therapeutic in vivo genome editing for the treatment of a multitude of human gene-based diseases.


Subject(s)
Dependovirus/genetics , Gene Editing , Hematopoietic Stem Cells/metabolism , Homologous Recombination , BRCA2 Protein/physiology , Genetic Vectors , Humans , Interleukin Receptor Common gamma Subunit/genetics , K562 Cells
2.
J Biol Chem ; 286(47): 40608-13, 2011 Nov 25.
Article in English | MEDLINE | ID: mdl-21965667

ABSTRACT

Fcγ receptors (FcγRs) play critical roles in humoral and cellular immune responses through interactions with the Fc region of immunoglobulin G (IgG). Among them, FcγRI is the only high affinity receptor for IgG and thus is a potential target for immunotherapy. Here we report the first crystal structure of an FcγRI with all three extracellular Ig-like domains (designated as D1, D2, and D3). The structure shows that, first, FcγRI has an acute D1-D2 hinge angle similar to that of FcεRI but much smaller than those observed in the low affinity Fcγ receptors. Second, the D3 domain of FcγRI is positioned away from the putative IgG binding site on the receptor and is thus unlikely to make direct contacts with Fc. Third, the replacement of FcγRIII FG-loop ((171)LVGSKNV(177)) with that of FcγRI ((171)MGKHRY(176)) resulted in a 15-fold increase in IgG(1) binding affinity, whereas a valine insertion in the FcγRI FG-loop ((171)MVGKHRY(177)) abolished the affinity enhancement. Thus, the FcγRI FG-loop with its conserved one-residue deletion is critical to the high affinity IgG binding. The structural results support FcγRI binding to IgG in a similar mode as its low affinity counterparts. Taken together, our study suggests a molecular mechanism for the high affinity IgG recognition by FcγRI and provides a structural basis for understanding its physiological function and its therapeutic implication in treating autoimmune diseases.


Subject(s)
Immunoglobulin G/metabolism , Receptors, IgG/chemistry , Receptors, IgG/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cricetulus , Crystallography, X-Ray , Humans , Immunoglobulin Fc Fragments/metabolism , Models, Molecular , Molecular Sequence Data , N-Acetylneuraminic Acid/metabolism , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Substrate Specificity
3.
Mol Ther Methods Clin Dev ; 26: 224-238, 2022 Sep 08.
Article in English | MEDLINE | ID: mdl-35859693

ABSTRACT

Adeno-associated viruses derived from human hematopoietic stem cells (AAVHSCs) are naturally occurring AAVs. Fifteen AAVHSCs have demonstrated broad biodistribution while displaying differences in transduction. We examine the structure-function relationships of these natural amino acid variations on cellular binding. We demonstrate that AAVHSC16 is the only AAVHSC that does not preferentially bind to terminal galactose. AAVHSC16 contains two unique amino acids, 501I and 706C, compared with other AAVHSCs. Through mutagenesis, we determined that residue 501 contributes to the lack of galactose binding. Structural analysis revealed that residue 501 is in proximity to the galactose binding pocket, hence confirming its functional role in galactose binding. Biodistribution analysis of AAVHSC16 indicated significantly less liver tropism in mice and non-human primates compared with other clade F members, likely associated with overall binding differences observed in vitro. AAVHSC16 maintained robust tropism to other key tissues in the peripheral and central nervous systems after intravenous injection, including to the brain, heart, and gastrocnemius. Importantly, AAVHSC16 did not induce elevated liver enzyme levels in non-human primates after intravenous injection at high doses. The unique glycan binding and tropism of AAVHSC16 makes this naturally occurring capsid an attractive candidate for therapies requiring less liver tropism while maintaining broad biodistribution.

4.
J Immunol ; 182(11): 7272-9, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19454724

ABSTRACT

Binding of immune complexes to cellular FcgammaRs can promote cell activation and inflammation. In previous studies, a recombinant human (rh) soluble FcgammaR, rh-FcgammaRIA (CD64A), was shown to block inflammation in passive transfer models of immune complex-mediated disease. To assess whether rh-FcgammaRIA could block inflammation in a T cell- and B cell-dependent model of immune complex-mediated disease, the efficacy of rh-FcgammaRIA in collagen-induced arthritis was evaluated. Mice with established arthritis were treated with a single s.c. injection of rh-FcgammaRIA (0.2-2.0 mg/dose) given every other day for 11 days. Relative to mice injected with vehicle alone, mice treated with rh-FcgammaRIA exhibited lower serum concentrations of IL-6, anti-type II collagen Abs, and total IgG2a. These changes were correlated with lower levels of paw swelling and joint damage in the rh-FcgammaRIA-treated mice and occurred in the presence of a significant murine Ab response to rh-FcgammaRIA. Comparison of the serum rh-FcgammaRIA concentration vs time profiles for rh-FcgammaRIA administered at two dose levels by i.v. and s.c. injection revealed that the bioavailabilty of s.c. administered rh-FcgammaRIA was 27-37%. Taken together, these data show that rh-FcgammaRIA is an effective inhibitor of inflammation in a model of established arthritis in mice.


Subject(s)
Arthritis/drug therapy , Inflammation/drug therapy , Receptors, IgG/administration & dosage , Animals , Antibodies/blood , Antibody Formation , Arthritis/chemically induced , Arthritis/pathology , Collagen/adverse effects , Collagen/immunology , Humans , Immunoglobulin G/blood , Interleukin-6/blood , Mice , Pharmacokinetics , Receptors, IgG/therapeutic use , Recombinant Proteins , Solubility , Treatment Outcome
5.
PLoS One ; 15(5): e0233373, 2020.
Article in English | MEDLINE | ID: mdl-32453743

ABSTRACT

Targeted gene integration via precise homologous recombination (HR)-based gene editing has the potential to correct genetic diseases. AAV (adeno-associated virus) can mediate nuclease-free gene integration at a disease-causing locus. Therapeutic application of AAV gene integration requires quantitative molecular characterization of the edited sequence that overcome technical obstacles such as excess episomal vector genomes and lengthy homology arms. Here we describe a novel molecular methodology that utilizes quantitative next-generation sequencing to characterize AAV-mediated targeted insertion and detects the presence of unintended mutations. The methods described here quantify targeted insertion and query the entirety of the target locus for the presence of insertions, deletions, single nucleotide variants (SNVs) and integration of viral components such as inverted terminal repeats (ITR). Using a humanized liver murine model, we demonstrate that hematopoietic stem-cell derived AAVHSC15 mediates in vivo targeted gene integration into human chromosome 12 at the PAH (phenylalanine hydroxylase) locus at 6% frequency, with no sign of co-incident random mutations at or above a lower limit of detection of 0.5% and no ITR sequences at the integration sites. Furthermore, analysis of heterozygous variants across the targeted locus using the methods described shows a pattern of strand cross-over, supportive of an HR mechanism of gene integration with similar efficiencies across two different haplotypes. Rapid advances in the application of AAV-mediated nuclease-free target integration, or gene editing, as a new therapeutic modality requires precise understanding of the efficiency and the nature of the changes being introduced to the target genome at the molecular level. This work provides a framework to be applied to homologous recombination gene editing platforms for assessment of introduced and natural sequence variation across a target site.


Subject(s)
Dependovirus/physiology , Gene Editing/methods , Liver/chemistry , Phenylalanine Hydroxylase/genetics , Animals , Genetic Vectors/administration & dosage , HEK293 Cells , High-Throughput Nucleotide Sequencing , Humans , Mice , Models, Animal , Mutation , Sequence Analysis, DNA , Virus Integration
6.
Mol Ther Methods Clin Dev ; 17: 568-580, 2020 Jun 12.
Article in English | MEDLINE | ID: mdl-32258219

ABSTRACT

Phenylketonuria is an inborn error of metabolism caused by loss of function of the liver-expressed enzyme phenylalanine hydroxylase and is characterized by elevated systemic phenylalanine levels that are neurotoxic. Current therapies do not address the underlying genetic disease or restore the natural metabolic pathway resulting in the conversion of phenylalanine to tyrosine. A family of hepatotropic clade F adeno-associated viruses (AAVs) was isolated from human CD34+ hematopoietic stem cells (HSCs) and one (AAVHSC15) was utilized to deliver a vector to correct the phenylketonuria phenotype in Pahenu2 mice. The AAVHSC15 vector containing a codon-optimized form of the human phenylalanine hydroxylase cDNA was administered as a single intravenous dose to Pahenu2 mice maintained on a phenylalanine-containing normal chow diet. Optimization of the transgene resulted in a vector that produced a sustained reduction in serum phenylalanine and normalized tyrosine levels for the lifespan of Pahenu2 mice. Brain levels of phenylalanine and the downstream serotonin metabolite 5-hydroxyindoleacetic acid were restored. In addition, the coat color of treated mice darkened following treatment, indicating restoration of the phenylalanine metabolic pathway. Taken together, these data support the potential of an AAVHSC15-based gene therapy as an investigational therapeutic for phenylketonuria patients.

7.
PLoS One ; 14(11): e0225582, 2019.
Article in English | MEDLINE | ID: mdl-31770409

ABSTRACT

The biodistribution of AAVHSC7, AAVHSC15, and AAVHSC17 following systemic delivery was assessed in cynomolgus macaques (Macaca fascicularis). Animals received a single intravenous (IV) injection of a self-complementary AAVHSC-enhanced green fluorescent protein (eGFP) vector and tissues were harvested at two weeks post-dose for anti-eGFP immunohistochemistry and vector genome analyses. IV delivery of AAVHSC vectors produced widespread distribution of eGFP staining in glial cells throughout the central nervous system, with the highest levels seen in the pons and lateral geniculate nuclei (LGN). eGFP-positive neurons were also observed throughout the central and peripheral nervous systems for all three AAVHSC vectors including brain, spinal cord, and dorsal root ganglia (DRG) with staining evident in neuronal cell bodies, axons and dendritic arborizations. Co-labeling of sections from brain, spinal cord, and DRG with anti-eGFP antibodies and cell-specific markers confirmed eGFP-staining in neurons and glia, including protoplasmic and fibrous astrocytes and oligodendrocytes. For all capsids tested, 50 to 70% of glial cells (S100-ß+) and on average 8% of neurons (NeuroTrace+) in the LGN were positive for eGFP expression. In the DRG, 45 to 62% of neurons and 8 to 12% of satellite cells were eGFP-positive for the capsids tested. eGFP staining was also observed in peripheral tissues with abundant staining in hepatocytes, skeletal- and cardio-myocytes and in acinar cells of the pancreas. Biodistribution of AAVHSC vector genomes in the central and peripheral organs generally correlated with eGFP staining and were highest in the liver for all AAVHSC vectors tested. These data demonstrate that AAVHSCs have broad tissue tropism and cross the blood-nerve and blood-brain-barriers following systemic delivery in nonhuman primates, making them suitable gene editing or gene transfer vectors for therapeutic application in human genetic diseases.


Subject(s)
Blood-Brain Barrier/metabolism , Central Nervous System/metabolism , Dependovirus/genetics , Genetic Vectors/metabolism , Administration, Intravenous , Animals , Ganglia, Spinal/metabolism , Genetic Therapy/methods , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Immunohistochemistry , Macaca , Neuroglia/metabolism , Neurons/metabolism , Tissue Distribution
8.
Hum Gene Ther Clin Dev ; 29(1): 60-67, 2018 03.
Article in English | MEDLINE | ID: mdl-29624457

ABSTRACT

To assess the therapeutic utility of AAVHSC15 and AAVHSC17, two recently described Clade F adeno-associated viruses (AAVs), the seroprevalence of neutralizing antibodies (NAbs) to these AAVs was assessed in a representative human population and compared to that of AAV9. NAb levels were measured in 100 unique human sera of different races (34, Black, 33 Caucasian, and 33 Hispanic) and sex (49% female, 51% male) collected within the United States. Fifty-six sera were tested in Huh7 cells and 44 sera were tested in 2V6.11 cells with vectors packaged with either a CMV-promoter upstream of LacZ or a CBA-promoter upstream of Firefly Luciferase, respectively. For AAVHSC15, AAVHSC17, and AAV9, 24/100 (24%), 21/100 (21%), and 17/100 (17%), respectively, of all sera tested were seropositive for NAbs using 50% inhibition of cellular transduction at a 1/16 dilution of serum as cutoff for seropositivity. Only 6% of positive sera had titers of 1/150 to 1/340, indicating that the majority of positive sera were of low titer. Significant cross-reactivity of NAbs across all three AAV serotypes was observed. These data show that approximately 80% of humans evaluated were seronegative for pre-existing NAbs to the AAV serotypes tested, suggesting that the vast majority of human subjects would be amenable to therapeutic intervention with Clade F AAVs.


Subject(s)
Antibodies, Neutralizing/immunology , Dependovirus/immunology , Racial Groups , Serogroup , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/genetics , Cell Line , Cell Line, Tumor , Dependovirus/genetics , Genetic Vectors/immunology , Humans , Seroepidemiologic Studies , United States
9.
Mol Endocrinol ; 20(2): 414-25, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16210345

ABSTRACT

Corticotroph-derived glycoprotein hormone (CGH), also referred to as thyrostimulin, is a noncovalent heterodimer of glycoprotein hormone alpha 2 (GPHA2) and glycoprotein hormone beta 5 (GPHB5). Here, we demonstrate that both subunits of CGH are expressed in the corticotroph cells of the human anterior pituitary, as well as in skin, retina, and testis. CGH activates the TSH receptor (TSHR); (125)I-CGH binding to cells expressing TSHR is saturable, specific, and of high affinity. In competition studies, unlabeled CGH is a potent competitor for (125)I-TSH binding, whereas unlabeled TSH does not compete for (125)I-CGH binding. Binding and competition analyses are consistent with the presence of two binding sites on the TSHR transfected baby hamster kidney cells, one that can interact with either TSH or CGH, and another that binds CGH alone. Transgenic overexpression of GPHB5 in mice produces elevations in serum T(4) levels, reductions in body weight, and proptosis. However, neither transgenic overexpression of GPHA2 nor deletion of GPHB5 produces an overt phenotype in mice. In vivo administration of CGH to mice produces a dose-dependent hyperthyroid phenotype including elevation of T(4) and hypertrophy of cells within the inner adrenal cortex. However, the distinctive expression patterns and binding characteristics of CGH suggest that it has endogenous biological roles that are discrete from those of TSH.


Subject(s)
Glycoproteins/metabolism , Receptors, Thyrotropin/metabolism , Animals , Binding Sites , Binding, Competitive , CHO Cells , Cricetinae , Cricetulus , Glycoproteins/analysis , Glycoproteins/genetics , Glycoproteins/pharmacology , Humans , Hypertrophy , Male , Mice , Mice, Transgenic , Peptide Hormones/analysis , Peptide Hormones/metabolism , Pituitary Gland, Anterior/chemistry , Pituitary Gland, Anterior/metabolism , Retina/chemistry , Retina/metabolism , Skin/chemistry , Skin/metabolism , Testis/chemistry , Testis/metabolism , Thyroid Gland/drug effects , Thyroid Gland/pathology , Thyroxine/blood , Tissue Distribution
10.
Stroke ; 34(6): 1507-12, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12738892

ABSTRACT

BACKGROUND AND PURPOSE: Fibroblast growth factor 18 (FGF18) is expressed in rodent brain and is a trophic factor for neuron-derived cells in culture. The purpose of the present study was to evaluate whether FGF18 was neuroprotective in a rat model of cerebral ischemia and to compare the results with those obtained with FGF2. METHODS: Cerebral ischemia was produced in rats by a transient 2-hour occlusion of the middle cerebral artery (MCAo) with an intraluminal filament followed by 22-hour reperfusion. Starting 15 minutes after MCAo, FGF18 or FGF2 was administered by a 3-hour intravenous infusion. Infarct volumes and behavioral deficits were measured 24 hours after MCAo. RESULTS: Infusion of FGF18 produced dose-dependent reductions in infarct volumes and improvements in tests of reference and working memory, motor ability, and exploratory behavior. FGF18 was more efficacious than FGF2 on virtually all measures examined. The reductions in infarct volume and behavioral deficit were associated with FGF-mediated increases in regional cerebral blood flow. CONCLUSIONS: These results demonstrate that FGF18 is an effective neuroprotective agent in a rat model of transient MCAo.


Subject(s)
Behavior, Animal/drug effects , Cerebral Infarction/prevention & control , Fibroblast Growth Factors/administration & dosage , Infarction, Middle Cerebral Artery/drug therapy , Ischemic Attack, Transient/drug therapy , Animals , Blood Flow Velocity/drug effects , Cerebral Infarction/etiology , Cerebral Infarction/pathology , Cerebrovascular Circulation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Fibroblast Growth Factor 2/administration & dosage , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/physiopathology , Infusions, Intravenous , Ischemic Attack, Transient/complications , Ischemic Attack, Transient/physiopathology , Male , Memory/drug effects , Motor Activity/drug effects , Neuroprotective Agents/administration & dosage , Rats , Rats, Sprague-Dawley , Treatment Outcome
11.
J Cereb Blood Flow Metab ; 24(1): 114-23, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14688623

ABSTRACT

To assess the time window for fibroblast growth factor-18 (FGF18)-mediated neuroprotection, FGF18 was administered by intravenous infusion at various times after transient occlusion of the middle cerebral artery (MCAO) in rats. Vehicle or FGF18 (100 microg x kg(-1) x h(-1)) was infused at 0.25, 0.5, 1.0, 2.0, 4.0, or 8.0 hours after MCAO with infarct volumes and behavioral deficits measured at 24.0 hours after MCAO. A separate group of animals received the infusions 24 hours after MCAO with endpoints measured at 48 hours after MCAO. Infusion of FGF18 reduced infarct volumes and improved scores in tests of reference and working memory, motor ability, and exploratory behavior. FGF18 was most efficacious when infused within 2 hours after MCAO. Significant reductions in infarct volumes and reductions in deficits of reference memory and motor activity were also observed with FGF18 infused 24 hours after MCAO. Measurements taken at infusion times before 2 hours after MCAO showed that regional cerebral blood flow was increased by FGF18. Administration of vehicle or FGF18 had no significant effect on mean arterial blood pressure, heart rate, brain temperature, blood pH, Pco2, or Po2. These results demonstrate that FGF18 is an effective neuroprotective agent when administered early after transient MCAO in rats. Efficacy observed with infusions at later times suggests an expanded time window for FGF18-mediated neuroprotection.


Subject(s)
Fibroblast Growth Factors/pharmacology , Middle Cerebral Artery/physiology , Neuroprotective Agents , Animals , Behavior, Animal/physiology , Cerebrovascular Circulation , Escherichia coli/metabolism , Exploratory Behavior/physiology , Fibroblast Growth Factors/biosynthesis , Humans , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/prevention & control , Infarction, Middle Cerebral Artery/psychology , Infusions, Intravenous , Male , Maze Learning/drug effects , Memory, Short-Term/physiology , Motor Activity/physiology , Psychomotor Performance/physiology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/biosynthesis , Recombinant Proteins/pharmacology
12.
Protein Eng Des Sel ; 23(4): 299-309, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20150179

ABSTRACT

A recombinant soluble version of the human high-affinity receptor for IgG, rh-FcgammaRIA or CD64A, was expressed in mammalian cells and purified from their conditioned media. As assessed by circular dichroism, size exclusion chromatography and dynamic light scattering, incubation of rh-FcgammaRIA at 37 degrees C resulted in time-dependent formation of soluble aggregates caused by protein unfolding and loss of native structure. Aggregate formation was irreversible, temperature-dependent and was independent of rh-FcgammaRIA concentration. Aggregated rh-FcgammaRIA lost its ability to inhibit immune complex precipitation and failed to bind to IgG-Sepharose. Addition of human IgG1 to rh-FcgammaRIA prior to incubation at 37 degrees C blocked the formation of rh-FcgammaRIA aggregates. Production of soluble monomeric rh-FcgammaRIA was limited by aggregate formation during cell culture. Substitution of the membrane distal D1 Ig domain of FcgammaRIA with the D1 Ig domain of FcgammaRIIIA or CD16A resulted in a chimeric receptor, FcgammaR3A1A, with enhanced temperature stability. Relative to native rh-FcgammaRIA, FcgammaR3A1A exhibited less aggregation in Chinese hamster ovary cell-conditioned media or when purified receptor was incubated for up to 24 h at 37 degrees C. Both receptors bound to immobilized human IgG1 with high affinity and were equipotent at blockade of immune complex-mediated cytokine production from cultured mast cells. Equivalent dose-dependent reductions in edema and neutrophil infiltration in the cutaneous Arthus reaction in mice were noted for rh-FcgammaRIA and FcgammaR3A1A. These data demonstrate that the D1 Ig domains of FcgammaRIA and FcgammaRIIIA are functionally interchangeable and further suggest that the chimeric receptor FcgammaR3A1A is an effective inhibitor of type III hypersensitivity in mice.


Subject(s)
Receptors, IgG/chemistry , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Mice , Mice, Inbred BALB C , Protein Structure, Tertiary , Receptors, IgG/immunology , Receptors, IgG/metabolism
13.
Arthritis Res Ther ; 12(2): R48, 2010.
Article in English | MEDLINE | ID: mdl-20302641

ABSTRACT

INTRODUCTION: B-lymphocyte stimulator (BLyS) and a proliferation-inducing ligand (APRIL) are members of the tumor necrosis factor (TNF) family that regulate B-cell maturation, survival, and function. They are overexpressed in a variety of autoimmune diseases and reportedly exist in vivo not only as homotrimers, but also as BLyS/APRIL heterotrimers. METHODS: A proprietary N-terminal trimerization domain was used to produce recombinant BLyS/APRIL heterotrimers. Heterotrimer biologic activity was compared with that of BLyS and APRIL in a 4-hour signaling assay by using transmembrane activator and CAML interactor (TACI)-transfected Jurkat cells and in a 4-day primary human B-cell proliferation assay. A bead-based immunoassay was developed to quantify native heterotrimers in human sera from healthy donors (n = 89) and patients with systemic lupus erythematosus (SLE; n = 89) or rheumatoid arthritis (RA; n = 30). Heterotrimer levels were compared with BLyS and APRIL homotrimer levels in a subset of these samples. RESULTS: The recombinant heterotrimers consisted mostly of one BLyS and two APRIL molecules. Heterotrimer signaling did not show any significant difference compared with APRIL in the TACI-Jurkat assay. Heterotrimers were less-potent inducers of B-cell proliferation than were homotrimeric BLyS or APRIL (EC(50), nMol/L: BLyS, 0.02; APRIL, 0.17; heterotrimers, 4.06). The soluble receptor fusion proteins atacicept and B-cell maturation antigen (BCMA)-immunoglobulin (Ig) neutralized the activity of BLyS, APRIL, and heterotrimers in both cellular assays, whereas B-cell activating factor belonging to the TNF family receptor (BAFF-R)-Ig neutralized only the activity of BLyS. In human sera, significantly more patients with SLE had detectable BLyS (67% versus 18%; P < 0.0001), APRIL (38% versus 3%; P < 0.0002), and heterotrimer (27% versus 8%; P = 0.0013) levels compared with healthy donors. Significantly more patients with RA had detectable APRIL, but not BLyS or heterotrimer, levels compared with healthy donors (83% versus 3%; P < 0.0001). Heterotrimer levels weakly correlated with BLyS, but not APRIL, levels. CONCLUSIONS: Recombinant BLyS/APRIL heterotrimers have biologic activity and are inhibited by atacicept and BCMA-Ig, but not by BAFF-R-Ig. A novel immunoassay demonstrated that native BLyS/APRIL heterotrimers, as well as BLyS and APRIL homotrimers, are elevated in patients with autoimmune diseases.


Subject(s)
Autoimmune Diseases/blood , B-Cell Activating Factor/blood , B-Cell Maturation Antigen/pharmacology , Cell Proliferation/drug effects , Recombinant Fusion Proteins/pharmacology , Tumor Necrosis Factor Ligand Superfamily Member 13/blood , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Humans , Interleukin-4/pharmacology , Jurkat Cells , Lymphocyte Activation/drug effects , Protein Multimerization , Recombinant Proteins
14.
J Immunol ; 180(1): 580-9, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18097060

ABSTRACT

Binding of Ag-Ab immune complexes to cellular FcgammaR promotes cell activation, release of inflammatory mediators, and tissue destruction characteristic of autoimmune disease. To evaluate whether a soluble FcgammaR could block the proinflammatory effects of immune complexes, recombinant human (rh) versions of FcgammaRIA, FcgammaRIIA, and FcgammaRIIIA were prepared. Binding of rh-FcgammaRIA to IgG was of high affinity (KD=1.7x10(-10) M), whereas rh-FcgammaRIIA and rh-FcgammaRIIIA bound with low affinity (KD=0.6-1.9x10(-6) M). All rh-FcgammaR reduced immune complex precipitation, blocked complement-mediated lysis of Ab-sensitized RBC, and inhibited immune complex-mediated production of IL-6, IL-13, MCP-1, and TNF-alpha by cultured mast cells. Local or systemic delivery only of rh-FcgammaRIA, however, reduced edema and neutrophil infiltration in the cutaneous Arthus reaction in mice. 125I-labeled rh-FcgammaRIA was cleared from mouse blood with a rapid distribution phase followed by a slow elimination phase with a t1/2gamma of approximately 130 h. The highest percentage of injected radioactivity accumulated in blood approximately liver approximately carcass>kidney. s.c. dosing of rh-FcgammaRIA resulted in lower serum levels of inflammatory cytokines and prevented paw swelling and joint damage in a murine model of collagen Ab-induced arthritis. These data demonstrate that rh-FcgammaRIA is an effective inhibitor of type III hypersensitivity.


Subject(s)
Immune Complex Diseases/drug therapy , Receptors, IgG/therapeutic use , Animals , Antigen-Antibody Complex/drug effects , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Arthus Reaction/drug therapy , Arthus Reaction/pathology , Complement System Proteins/immunology , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Humans , Immune Complex Diseases/pathology , Immunoglobulin G/metabolism , Mast Cells/immunology , Mice , Receptors, IgG/biosynthesis
15.
J Immunol ; 179(8): 5462-73, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17911633

ABSTRACT

The proinflammatory cytokines IL-17A and IL-17F have a high degree of sequence similarity and share many biological properties. Both have been implicated as factors contributing to the progression of inflammatory and autoimmune diseases. Moreover, reagents that neutralize IL-17A significantly ameliorate disease severity in several mouse models of human disease. IL-17A mediates its effects through interaction with its cognate receptor, the IL-17 receptor (IL-17RA). We report here that the IL-17RA-related molecule, IL-17RC is the receptor for IL-17F. Notably, both IL-17A and IL-17F bind to IL-17RC with high affinity, leading us to suggest that a soluble form of this molecule may serve as an effective therapeutic antagonist of IL-17A and IL-17F. We generated a soluble form of IL-17RC and demonstrate that it effectively blocks binding of both IL-17A and IL-17F, and that it inhibits signaling in response to these cytokines. Collectively, our work indicates that IL-17RC functions as a receptor for both IL-17A and IL-17F and that a soluble version of this protein should be an effective antagonist of IL-17A and IL-17F mediated inflammatory diseases.


Subject(s)
Interleukin-17/metabolism , Receptors, Interleukin-17/metabolism , Alternative Splicing/immunology , Animals , Binding, Competitive/immunology , Cell Line , Cricetinae , Humans , Inflammation Mediators/metabolism , Inflammation Mediators/therapeutic use , Interleukin-17/antagonists & inhibitors , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Protein Binding/genetics , Protein Binding/immunology , Receptors, Interleukin-17/genetics , Receptors, Interleukin-17/therapeutic use , Species Specificity , Transfection
16.
Blood ; 107(2): 423-30, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16195328

ABSTRACT

CTRP-1 is a novel member of the C1qTNF-related protein family containing family characteristic collagen and TNF-like domains and shows marked expression in vascular wall tissue. We observed that recombinant human CTRP-1 specifically bound to fibrillar collagen and blocked collagen-induced platelet aggregation. CTRP-1 completely or partially prevented VWF and GPVI-Fc4 binding to collagen, respectively. However, GPVI-Fc4 failed to compete for the binding of CTRP-1 to collagen. CTRP-1 had no effects on alpha(2)beta(1) integrin I-domain binding to collagen. Using whole human blood under flow at low and high shear rates, CTRP-1 prevented platelets from accumulating on a collagen-coated surface but had no effects on "platelet-rolling" on a surface coated with VWF. These data suggest that CTRP-1 prevents collagen-induced platelet aggregation by specific blockade of VWF binding to collagen. By using the Folts vascular injury model in nonhuman primates (Macaca fascicularis), we were able to demonstrate that CTRP-1 can prevent platelet thrombosis in vivo. This effect was achieved in the absence of changes in activated-clotting time (ACT) and template cut bleeding times, suggesting that CTRP-1 has promising antiplatelet thrombotic activity and most likely acts by pacifying the thrombogenic site of vascular injury.


Subject(s)
Collagen Type I/pharmacology , Endothelium, Vascular/metabolism , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation , Thrombosis/physiopathology , von Willebrand Factor/metabolism , Animals , Endothelium, Vascular/injuries , Endothelium, Vascular/physiopathology , Humans , Integrin alpha2beta1/metabolism , Macaca fascicularis , Platelet Adhesiveness , Platelet Membrane Glycoproteins/genetics , Platelet Membrane Glycoproteins/metabolism , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/pharmacology , Thrombosis/prevention & control , von Willebrand Factor/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL