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










Publication year range
1.
Nat Med ; 25(2): 229-233, 2019 02.
Article in English | MEDLINE | ID: mdl-30664785

ABSTRACT

Leber congenital amaurosis type 10 is a severe retinal dystrophy caused by mutations in the CEP290 gene1,2. We developed EDIT-101, a candidate genome-editing therapeutic, to remove the aberrant splice donor created by the IVS26 mutation in the CEP290 gene and restore normal CEP290 expression. Key to this therapeutic, we identified a pair of Staphylococcus aureus Cas9 guide RNAs that were highly active and specific to the human CEP290 target sequence. In vitro experiments in human cells and retinal explants demonstrated the molecular mechanism of action and nuclease specificity. Subretinal delivery of EDIT-101 in humanized CEP290 mice showed rapid and sustained CEP290 gene editing. A comparable surrogate non-human primate (NHP) vector also achieved productive editing of the NHP CEP290 gene at levels that met the target therapeutic threshold, and demonstrated the ability of CRISPR/Cas9 to edit somatic primate cells in vivo. These results support further development of EDIT-101 for LCA10 and additional CRISPR-based medicines for other inherited retinal disorders.


Subject(s)
Gene Editing , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/physiopathology , Animals , Cell Line , Gene Knock-In Techniques , Humans , Mice , Primates , Reproducibility of Results , Vision, Ocular
2.
Hum Gene Ther ; 30(5): 571-589, 2019 05.
Article in English | MEDLINE | ID: mdl-30358434

ABSTRACT

Mutations in GUCY2D, the gene encoding retinal guanylate cyclase-1 (retGC1), are the leading cause of autosomal dominant cone-rod dystrophy (CORD6). Significant progress toward clinical application of gene replacement therapy for Leber congenital amaurosis (LCA) due to recessive mutations in GUCY2D (LCA1) has been made, but a different approach is needed to treat CORD6 where gain of function mutations cause dysfunction and dystrophy. The CRISPR/Cas9 gene editing system efficiently disrupts genes at desired loci, enabling complete gene knockout or homology directed repair. Here, adeno-associated virus (AAV)-delivered CRISPR/Cas9 was used specifically to edit/disrupt this gene's early coding sequence in mouse and macaque photoreceptors in vivo, thereby knocking out retGC1 expression and demonstrably altering retinal function and structure. Neither preexisting nor induced Cas9-specific T-cell responses resulted in ocular inflammation in macaques, nor did it limit GUCY2D editing. The results show, for the first time, the ability to perform somatic gene editing in primates using AAV-CRISPR/Cas9 and demonstrate the viability this approach for treating inherited retinal diseases in general and CORD6 in particular.


Subject(s)
CRISPR-Cas Systems , Dependovirus/genetics , Gene Editing , Guanylate Cyclase/genetics , Receptors, Cell Surface/genetics , Retina/metabolism , Animals , Base Sequence , Electroretinography , Genes, Reporter , Genetic Vectors/genetics , Guanylate Cyclase/metabolism , Macaca , Mice , Mice, Knockout , Molecular Imaging/methods , Promoter Regions, Genetic , RNA, Guide, Kinetoplastida/chemistry , RNA, Guide, Kinetoplastida/genetics , Receptors, Cell Surface/metabolism , Retina/pathology
3.
Toxicol Pathol ; 43(4): 519-29, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25274659

ABSTRACT

Batracylin (NSC-320846) is a dual inhibitor of DNA topoisomerases I and II. Batracylin advanced as an anticancer agent to Phase I clinical trials where dose limiting hemorrhagic cystitis (bladder inflammation and bleeding) was observed. To further investigate batracylin's mechanism of toxicity, studies were conducted in Fischer 344 rats. Once daily oral administration of 16 or 32 mg/kg batracylin to rats for 4 days caused overt toxicity. Abnormal clinical observations and adverse effects on clinical pathology, urinalysis, and histology indicated acute renal damage and urothelial damage and bone marrow dysfunction. Scanning electron microscopy revealed sloughing of the superficial and intermediate urothelial layers. DNA damage was evident in kidney and bone marrow as indicated by histone γ-H2AX immunofluorescence. After a single oral administration of 16 or 32 mg/kg, the majority of batracylin was converted to N-acetylbatracylin (NAB) with a half-life of 4 hr to 11 hr. Mesna (Mesnex™), a drug known to reduce the incidence of hemorrhagic cystitis induced by ifosfamide or cyclophosphamide, was administered to rats prior to batracylin, but did not alleviate batracylin-induced bladder and renal toxicity. These findings suggest that batracylin results in DNA damage-based mechanisms of toxicity and not an acrolein-based mechanism of toxicity as occurs after ifosfamide or cyclophosphamide administration.


Subject(s)
Kidney Neoplasms/chemically induced , Quinazolines/toxicity , Urinary Bladder Neoplasms/metabolism , Animals , Biomarkers, Tumor/analysis , Body Weight/drug effects , Female , Glycosuria/chemically induced , Histones/metabolism , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Male , Mesna/pharmacology , Phosphoproteins/metabolism , Quinazolines/pharmacokinetics , Random Allocation , Rats , Urinary Bladder Neoplasms/chemically induced , Urinary Bladder Neoplasms/pathology
4.
Mol Genet Metab ; 111(2): 152-62, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24388731

ABSTRACT

Clinical, epidemiological and experimental studies confirm a connection between the common degenerative movement disorder Parkinson's disease (PD) that affects over 1 million individuals, and Gaucher disease, the most prevalent lysosomal storage disorder. Recently, human imaging studies have implicated impaired striatal dopaminergic neurotransmission in early PD pathogenesis in the context of Gaucher disease mutations, but the underlying mechanisms have yet to be characterized. In this report we describe and characterize two novel long-lived transgenic mouse models of Gba deficiency, along with a subchronic conduritol-ß-epoxide (CBE) exposure paradigm. All three murine models revealed striking glial activation within nigrostriatal pathways, accompanied by abnormal α-synuclein accumulation. Importantly, the CBE-induced, pharmacological Gaucher mouse model replicated this change in dopamine neurotransmission, revealing a markedly reduced evoked striatal dopamine release (approximately 2-fold) that indicates synaptic dysfunction. Other changes in synaptic plasticity markers, including microRNA profile and a 24.9% reduction in post-synaptic density size, were concomitant with diminished evoked dopamine release following CBE exposure. These studies afford new insights into the mechanisms underlying the Parkinson's-Gaucher disease connection, and into the physiological impact of related abnormal α-synuclein accumulation and neuroinflammation on nigrostriatal dopaminergic neurotransmission.


Subject(s)
Corpus Striatum/pathology , Gaucher Disease/pathology , Glucosylceramidase , Parkinson Disease/pathology , Synapses/pathology , alpha-Synuclein/metabolism , Animals , Corpus Striatum/enzymology , Corpus Striatum/physiopathology , Disease Models, Animal , Dopamine/metabolism , Evoked Potentials, Motor , Female , Gaucher Disease/enzymology , Gaucher Disease/genetics , Gaucher Disease/physiopathology , Humans , Inflammation , Inositol/administration & dosage , Inositol/analogs & derivatives , Male , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Mutation , Neuronal Plasticity , Parkinson Disease/enzymology , Parkinson Disease/genetics , Parkinson Disease/physiopathology , Synapses/enzymology , Synaptic Transmission , alpha-Synuclein/genetics
5.
Eur J Pharmacol ; 683(1-3): 285-93, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22449384

ABSTRACT

Inflammatory bowel diseases, primarily Crohn's disease and ulcerative colitis, are chronic inflammatory disorders of the gastrointestinal tract with unknown etiology. The majority of current therapeutic agents focus on controlling proinflammatory molecules. The neuropeptide nociceptin/orphanin FQ (N/OFQ) has been described as a potential immunomodulator for inflammatory bowel diseases. In this study, we asked whether the small molecule N/OFQ antagonist (-)-cis-1-methyl-7-[[4-(2,6-dichlorophenyl)piperidin-1-yl]methyl]-6,7,8,9-tetrahydro-5H-benzocyclohepten-5-ol (SB612111) would inhibit the development of dextran sodium sulfate-induced colitis in C57BL/6 mice. Inhibition of the N/OFQ receptor (NOP) by SB612111 significantly ameliorated the clinical disease course in these animals, as indicated by reduced fecal bleeding, improved recovery from diarrhea and weight loss, and a reduction in histopathological alterations. In addition, the inflammatory response in the colon was diminished, as demonstrated by reduced cytokine protein and messenger RNA expression for CXCL1/keratinocyte-derived chemokine, interferon-γ, interleukin-1ß, interleukin-6, and tumor necrosis factor-α, some of which are known targets for the treatment of this devastating disease. Our results strongly support a role for the receptor-ligand pair NOP-N/OFQ in the pathogenesis of colitis. We conclude that inhibition of NOP receptors with small molecule inhibitors may constitute a novel, urgently needed approach for the treatment of inflammatory bowel diseases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Colitis/prevention & control , Colon/drug effects , Cycloheptanes/therapeutic use , Narcotic Antagonists , Opioid Peptides/antagonists & inhibitors , Piperidines/therapeutic use , Signal Transduction/drug effects , Animals , Colitis/immunology , Colitis/metabolism , Colitis/physiopathology , Colon/immunology , Colon/metabolism , Colon/pathology , Cytokines/genetics , Cytokines/metabolism , Dextran Sulfate , Diarrhea/etiology , Diarrhea/prevention & control , Female , Gastrointestinal Hemorrhage/etiology , Gastrointestinal Hemorrhage/prevention & control , Gene Expression Regulation/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy , RNA, Messenger/metabolism , Receptors, Opioid , Weight Loss/drug effects , Nociceptin Receptor , Nociceptin
6.
Biomacromolecules ; 13(3): 579-83, 2012 Mar 12.
Article in English | MEDLINE | ID: mdl-22304720

ABSTRACT

The nonapeptide hemopressin, which is derived from the α chain of hemoglobin, has been reported to exhibit inverse agonist activity against the CB1 receptor. Administration of this peptide in animal models led to decreased food intake and elicited hypotensive and antinociceptive effects. On the basis of hemopressin's potential in therapeutic applications and the lack of a structure-activity relationship study in literature, we aimed to determine the conformational features of hemopressin under physiological conditions. We conducted transmission electron microscopy experiments of hemopressin, revealing that it self-assembles into fibrils under aqueous conditions at pH 7.4. Circular dichroism and nuclear magnetic resonance experiments indicate that the peptide adopts a mostly extended ß-like structure, which may contribute to its self-assembly and fibril formation.


Subject(s)
Hemoglobins/chemistry , Hemoglobins/metabolism , Nanostructures , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Animals , Circular Dichroism , Hydrogen-Ion Concentration , Microscopy, Electron, Transmission , Models, Molecular , Rats , Receptor, Cannabinoid, CB1/agonists , Structure-Activity Relationship
7.
Int J Cancer ; 128(9): 2230-9, 2011 May 01.
Article in English | MEDLINE | ID: mdl-20635388

ABSTRACT

We found that adenosine 5'-monophosphate-activated protein kinase (AMPK), which is considered the "fuel sensor" of mammalian cells because it directly responds to the depletion of the fuel molecule ATP, is strongly activated by tumor-like hypoxia and glucose deprivation. We also observed abundant AMPK activity in tumor cells in vivo, using subcutaneous tumor xenografts prepared from cells transformed with oncogenic H-Ras. Such rapidly growing transplants of tumor cells, however, represent fully developed tumors that naturally contain energetically stressed microenvironments that can activate AMPK. Therefore, to investigate the induction of AMPK activity during experimental tumorigenesis, we used an established model of brain tumor (glioma) development in the offspring of rats exposed prenatally to the mutagen N-ethyl-N-nitrosourea. We observed that immunostaining for a specific readout of AMPK activity (AMPK-dependent phosphorylation of acetyl-CoA carboxylase) was prominent during N-ethyl-N-nitrosourea-initiated neurocarcinogenesis, from the occurrence of early hyperplasia (microtumors) to the emergence of large gliomas. Moreover, we observed that immunostaining for activating phosphorylation of AMPK correlated with the same stages of glioma development, notably in mitotic tumor cells in which the signal showed punctate as well as cytoplasmic patterns associated with spindle formation. Based on these observations, we propose that neurocarcinogenesis requires AMPK-dependent regulation of cellular energy metabolism.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Brain Neoplasms/enzymology , Cell Transformation, Neoplastic/metabolism , Glioma/enzymology , Acetyl-CoA Carboxylase/metabolism , Animals , Blotting, Western , Brain Neoplasms/chemically induced , Brain Neoplasms/pathology , Carcinogens/toxicity , Cell Transformation, Neoplastic/pathology , Ethylnitrosourea/toxicity , Fluorescent Antibody Technique , Glioma/chemically induced , Glioma/pathology , Immunohistochemistry , Neoplasm Staging , Phosphorylation , Rats , Rats, Sprague-Dawley
8.
Cancer Biol Ther ; 9(6): 469-76, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20087061

ABSTRACT

We previously identified metal-responsive transcription factor-1 (MTF-1) as a positive contributor to mouse fibrosarcoma growth through effects on cell survival, proliferation, tumor angiogenesis and extracellular matrix remodeling. In the present study, we investigated MTF-1 protein expression in human tissues by specific immunostaining of both normal and tumor tissue samples. Immunohistochemical (IHC) staining of a human tissue microarray (TMA), using a unique anti-human MTF-1 antibody, indicated constitutive MTF-1 expression in most normal tissues, with liver and testis displaying comparatively high levels of expression. Nevertheless, MTF-1 protein levels were found to be significantly elevated in diverse human tumor types, including breast, lung and cervical carcinomas. IHC analysis of a separate panel of full-size tissue sections of human breast cancers, including tumor and normal adjacent, surrounding tissue, confirmed and extended the results of the TMA analysis. Taken with our previous findings, this new study suggests a role for MTF-1 in human tumor development, growth or spread. Moreover, the study suggests that MTF-1 could be a novel therapeutic target that offers the opportunity to manipulate metal or redox homeostasis in tumor cells.


Subject(s)
DNA-Binding Proteins/metabolism , Neoplasms/metabolism , Transcription Factors/metabolism , Breast/chemistry , Breast/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , DNA-Binding Proteins/genetics , Female , Humans , Neoplasms/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Transcription Factors/genetics , Transcription Factor MTF-1
9.
Am J Pathol ; 171(1): 53-67, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17591953

ABSTRACT

Vascular endothelial growth factor-A (VEGF-A) has recently been recognized as an important neuroprotectant in the central nervous system. Given its position as an anti-angiogenic target in the treatment of human diseases, understanding the extent of VEGF's role in neural cell survival is paramount. Here, we used a model of ischemia-reperfusion injury and found that VEGF-A exposure resulted in a dose-dependent reduction in retinal neuron apoptosis. Although mechanistic studies suggested that VEGF-A-induced volumetric blood flow to the retina may be partially responsible for the neuroprotection, ex vivo retinal culture demonstrated a direct neuroprotective effect for VEGF-A. VEGF receptor-2 (VEGFR2) expression was detected in several neuronal cell layers of the retina, and functional analyses showed that VEGFR2 was involved in retinal neuroprotection. VEGF-A was also shown to be involved in the adaptive response to retinal ischemia. Ischemic preconditioning 24 hours before ischemia-reperfusion injury increased VEGF-A levels and substantially decreased the number of apoptotic retinal cells. The protective effect of ischemic preconditioning was reversed after VEGF-A inhibition. Finally, chronic inhibition of VEGF-A function in normal adult animals led to a significant loss of retinal ganglion cells yet had no observable effect on several vascular parameters. These findings have implications for both neural pathologies and ocular vascular diseases, such as diabetic retinopathy and age-related macular degeneration.


Subject(s)
Reperfusion Injury/metabolism , Retina/physiology , Vascular Endothelial Growth Factor A/physiology , Adult , Animals , Apoptosis , Blood Flow Velocity , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Macular Degeneration , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Organ Culture Techniques , Rats , Rats, Long-Evans , Reperfusion Injury/pathology , Retina/drug effects , Retinal Vessels/physiology , Vascular Endothelial Growth Factor Receptor-2/physiology
10.
Proc Natl Acad Sci U S A ; 103(45): 16770-5, 2006 Nov 07.
Article in English | MEDLINE | ID: mdl-17075074

ABSTRACT

Fenestrae are small pores in the endothelium of renal glomerular, gastrointestinal, and endocrine gland capillaries and are involved in the bidirectional exchange of molecules between blood and tissues. Although decades of studies have characterized fenestrae at the ultrastructural level, little is known on the mechanisms by which fenestrae form. We present the development of an in vitro assay in which rapid and abundant fenestra induction enables a detailed study of their biogenesis. Through the use of agents that stabilize or disassemble actin microfilaments, we show that actin microfilament remodeling is part of fenestra biogenesis in this model. Furthermore, by using a loss-of-function approach, we show that the diaphragm protein PV-1 is necessary for fenestral pore architecture and the ordered arrangement of fenestrae in sieve plates. Together, these data provide insight into the cell biology of fenestra formation and open up the future study of the fenestra to a combined morphological and biochemical analysis.


Subject(s)
Carrier Proteins/metabolism , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Membrane Proteins/metabolism , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Actins/metabolism , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Caveolin 1/metabolism , Cell Line , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Immunohistochemistry , In Vitro Techniques , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Microscopy, Electron , RNA, Small Interfering/genetics
11.
J Histochem Cytochem ; 52(7): 967-77, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15208363

ABSTRACT

Our objective in this study was to localize the corticotropin-releasing factor 2 receptor (CRF2R) in rodent and human skeletal muscle. We found CRF2R protein to be abundant in neural tissues in skeletal muscle, including large nerve fibers and bundles, neural tissue associated with mechanoreceptors, muscle spindles, and the Golgi tendon organ. CRF2R protein was also abundant in blood vessels in skeletal muscle. CRF2R protein was also observed, although with less abundance, in the endo/perimysial regions in skeletal muscle. The localization of the CRF2R to blood vessels is consistent with the CRF2R-mediated vascular phenomena observed previously, but the observation of CRF2R in neural tissue in skeletal muscle is a novel finding with an unknown function.


Subject(s)
Muscle, Skeletal/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Animals , Blood Vessels/metabolism , Female , Fluorescent Antibody Technique , Humans , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/blood supply , Muscle, Skeletal/innervation , Nerve Tissue/metabolism , Organ Specificity , Rats , Rats, Sprague-Dawley , Species Specificity
12.
Am J Physiol Endocrinol Metab ; 285(4): E889-98, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12959937

ABSTRACT

Two receptors activated by the corticotropin-releasing factor (CRF) family of peptides have been identified, the CRF 1 receptor (CRF1R) and the CRF 2 receptor (CRF2R). Of these, the CRF2R is expressed in skeletal muscle. To understand the role of the CRF2R in skeletal muscle, we utilized CRFR knockout mice and CRF2R-selective agonists to modulate nerve damage and corticosteroid- and disuse-induced skeletal muscle atrophy in mice. These analyses demonstrated that activation of the CRF2R decreased nerve damage and corticosteroid- and disuse-induced skeletal muscle mass and function loss. In addition, selective activation of the CRF2R increased nonatrophy skeletal muscle mass. Thus we describe for the first time a novel activity of the CRF2R, modulation of skeletal muscle mass.


Subject(s)
Muscle, Skeletal/physiology , Muscle, Skeletal/physiopathology , Receptors, Corticotropin-Releasing Hormone/deficiency , Receptors, Corticotropin-Releasing Hormone/physiology , Amphibian Proteins , Animals , Denervation , Dexamethasone , Female , Hindlimb , Male , Mice , Mice, Inbred C57BL , Muscle Contraction , Muscle, Skeletal/drug effects , Muscular Atrophy/chemically induced , Muscular Atrophy/physiopathology , Muscular Disorders, Atrophic/physiopathology , Organ Size/physiology , Peptide Hormones , Peptides/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Corticotropin-Releasing Hormone/agonists , Sciatic Nerve/surgery , Stress, Mechanical
13.
Nat Struct Biol ; 10(1): 38-44, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12469114

ABSTRACT

Angiopoietins are a recently discovered family of angiogenic factors that interact with the endothelial receptor tyrosine kinase Tie2, either as agonists (angiopoietin-1) or as context-dependent agonists/antagonists (angiopoietin-2). Here we show that angiopoietin-1 has a modular structure unlike any previously characterized growth factor. This modular structure consists of a receptor-binding domain, a dimerization motif and a superclustering motif that forms variable-sized multimers. Genetic engineering of precise multimers of the receptor-binding domain of angiopoietin-1, using surrogate multimerization motifs, reveals that tetramers are the minimal size required for activating endothelial Tie2 receptors. In contrast, engineered dimers can antagonize endothelial Tie2 receptors. Surprisingly, angiopoietin-2 has a modular structure and multimerization state similar to that of angiopoietin-1, and its antagonist activity seems to be a subtle property encoded in its receptor-binding domain.


Subject(s)
Angiopoietins/chemistry , Angiopoietins/metabolism , Receptor, TIE-2/metabolism , Amino Acid Motifs , Amino Acid Sequence , Angiopoietin-1/chemistry , Angiopoietin-1/genetics , Angiopoietin-1/metabolism , Angiopoietin-2/chemistry , Angiopoietin-2/genetics , Angiopoietin-2/metabolism , Angiopoietins/genetics , Animals , CHO Cells , Cricetinae , Cricetulus , Dimerization , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Models, Molecular , Phosphorylation , Protein Binding , Protein Engineering , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
14.
Biochemistry ; 41(37): 11091-8, 2002 Sep 17.
Article in English | MEDLINE | ID: mdl-12220173

ABSTRACT

Collagens have recently been identified as ligands for discoidin domain receptors (DDR1 and DDR2), generating an interest in studying the properties of binding of DDR to its ligand. We are interested in the interaction of DDR2 with collagen I because of its potential role in liver fibrosis. Our in vitro binding assay utilizes DDR2-Fc fusion proteins, which can be clustered (multimerized) by use of antibodies to form DDR2 complexes. Binding of DDR2 complexes to collagen I coated on plastic plates was established by a microplate-based assay using Eu(3+)-labeled proteins and time-resolved fluorometry. Clustering of the DDR2-Fc with antibody was found to be requisite for binding to collagen in vitro. Using atomic force microscopy (AFM) in an aqueous environment, we characterized the surface topographies of DDR2 complexes and collagen I, and investigated binding of this receptor-ligand pair. We were able to image and identify binding of DDR2 complexes onto individual molecules of triple-helical collagen and provide insight into the number and locations of binding sites on collagen I. In most cases, a single receptor complex bound to a single collagen molecule and there were preferred DDR2 binding sites on the collagen I triple helix. These data were validated by rotary-replication transmission electron microscopy (TEM) of glycerol-sprayed samples.


Subject(s)
Collagen Type I/chemistry , Collagen Type I/ultrastructure , Microscopy, Atomic Force/methods , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/ultrastructure , Receptors, Mitogen/chemistry , Receptors, Mitogen/ultrastructure , Animals , Binding Sites/genetics , Discoidin Domain Receptors , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin G/genetics , Ligands , Mice , Protein Binding/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Mitogen/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...