Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
PLoS One ; 18(10): e0286432, 2023.
Article in English | MEDLINE | ID: mdl-37862305

ABSTRACT

The prevailing concept is that gestational alloimmune liver disease (GALD) is caused by maternal antibodies targeting a currently unknown antigen on the liver of the fetus. This leads to deposition of complement on the fetal hepatocytes and death of the fetal hepatocytes and extensive liver injury. In many cases, the newborn dies. In subsequent pregnancies early treatment of the woman with intravenous immunoglobulin can be instituted, and the prognosis for the fetus will be excellent. Without treatment the prognosis can be severe. Crucial improvements of diagnosis require identification of the target antigen. For this identification, this work was based on two hypotheses: 1. The GALD antigen is exclusively expressed in the fetal liver during normal fetal life in all pregnancies; 2. The GALD antigen is an alloantigen expressed in the fetal liver with the woman being homozygous for the minor allele and the father being, most frequently, homozygous for the major allele. We used three different experimental approaches to identify the liver target antigen of maternal antibodies from women who had given birth to a baby with the clinical GALD diagnosis: 1. Immunoprecipitation of antigens from either a human liver cell line or human fetal livers by immunoprecipitation with maternal antibodies followed by mass spectrometry analysis of captured antigens; 2. Construction of a cDNA expression library from human fetal liver mRNA and screening about 1.3 million recombinants in Escherichia coli using antibodies from mothers of babies diagnosed with GALD; 3. Exome/genome sequencing of DNA from 26 presumably unrelated women who had previously given birth to a child with GALD with husband controls and supplementary HLA typing. In conclusion, using the three experimental approaches we did not identify the GALD target antigen and the exome/genome sequencing results did not support the hypothesis that the GALD antigen is an alloantigen, but the results do not yield basis for excluding that the antigen is exclusively expressed during fetal life., which is the hypothesis we favor.


Subject(s)
Digestive System Diseases , Fetal Diseases , Hemochromatosis , Infant, Newborn, Diseases , Liver Diseases , Thrombocytopenia, Neonatal Alloimmune , Child , Female , Humans , Infant, Newborn , Pregnancy , Hemochromatosis/diagnosis , Isoantigens , Liver Diseases/drug therapy
2.
J Biol Chem ; 295(7): 1781-1791, 2020 02 14.
Article in English | MEDLINE | ID: mdl-31819010

ABSTRACT

Iron is an essential nutrient for all living organisms. To acquire iron, many pathogens have developed elaborate systems to steal it from their hosts. The iron acquisition system in the opportunistic pathogen Staphylococcus aureus comprises nine proteins, called iron-regulated surface determinants (Isds). The Isd components enable S. aureus to extract heme from hemoglobin (Hb), transport it into the bacterial cytoplasm, and ultimately release iron from the porphyrin ring. IsdB and IsdH act as hemoglobin receptors and are known to actively extract heme from extracellular Hb. To limit microbial pathogenicity during infection, host organisms attempt to restrict the availability of nutrient metals at the host-pathogen interface. The human acute phase protein haptoglobin (Hp) protects the host from oxidative damage by clearing hemoglobin that has leaked from red blood cells and also restricts the availability of extracellular Hb-bound iron to invading pathogens. To investigate whether Hp serves an additional role in nutritional immunity through a direct inhibition of IsdH-mediated iron acquisition, here we measured heme extraction from the Hp-Hb complex by UV-visible spectroscopy and determined the crystal structure of the Hp-Hb-IsdH complex at 2.9 Å resolution. We found that Hp strongly inhibits IsdH-mediated heme extraction and that Hp binding prevents local unfolding of the Hb heme pocket, leaving IsdH unable to wrest the heme from Hb. Furthermore, we noted that the Hp-Hb binding appears to trap IsdH in an initial state before heme transfer. Our findings provide insights into Hp-mediated IsdH inhibition and the dynamics of IsdH-mediated heme extraction.


Subject(s)
Antigens, Bacterial/chemistry , Haptoglobins/chemistry , Host-Pathogen Interactions/genetics , Receptors, Cell Surface/chemistry , Staphylococcal Infections/genetics , Crystallography, X-Ray , Erythrocytes/metabolism , Erythrocytes/microbiology , Haptoglobins/genetics , Haptoglobins/ultrastructure , Heme/chemistry , Heme/genetics , Hemoglobins/chemistry , Hemoglobins/genetics , Humans , Iron/chemistry , Iron/metabolism , Protein Binding/genetics , Protein Conformation , Receptors, Cell Surface/antagonists & inhibitors , Staphylococcal Infections/blood , Staphylococcal Infections/microbiology , Staphylococcus aureus/chemistry , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity
3.
J Biol Chem ; 294(31): 11817-11828, 2019 08 02.
Article in English | MEDLINE | ID: mdl-31197037

ABSTRACT

The serine protease high-temperature requirement protein A1 (HtrA1) is associated with protein-misfolding disorders such as Alzheimer's disease and transforming growth factor ß-induced protein (TGFBIp)-linked corneal dystrophy. In this study, using several biochemical and biophysical approaches, including recombinant protein expression, LC-MS/MS and 2DE analyses, and thioflavin T (ThT) fluorescence assays for amyloid fibril detection, and FTIR assays, we investigated the role of HtrA1 both in normal TGFBIp turnover and in corneal amyloid formation. We show that HtrA1 can cleave WT TGFBIp but prefers amyloidogenic variants. Corneal TGFBIp is extensively processed in healthy people, resulting in C-terminal degradation products spanning the FAS1-4 domain of TGFBIp. We show here that HtrA1 cleaves the WT FAS1-4 domain only inefficiently, whereas the amyloidogenic FAS1-4 mutations transform this domain into a considerably better HTRA1 substrate. Moreover, HtrA1 cleavage of the mutant FAS1-4 domains generated peptides capable of forming in vitro amyloid aggregates. Significantly, these peptides have been previously identified in amyloid deposits in vivo, supporting the idea that HtrA1 is a causative agent for TGFBIp-associated amyloidosis in corneal dystrophy. In summary, our results indicate that TGFBIp is an HtrA1 substrate and that some mutations in the gene encoding TGFBIp cause aberrant HtrA1-mediated processing that results in amyloidogenesis in corneal dystrophies.


Subject(s)
Amyloid/metabolism , Extracellular Matrix Proteins/metabolism , High-Temperature Requirement A Serine Peptidase 1/metabolism , Transforming Growth Factor beta/metabolism , Aged, 80 and over , Chromatography, High Pressure Liquid , Cornea/metabolism , Corneal Diseases/metabolism , Corneal Diseases/pathology , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/genetics , High-Temperature Requirement A Serine Peptidase 1/genetics , Humans , Mutagenesis, Site-Directed , Peptides/analysis , Peptides/metabolism , Protein Domains , Protein Folding , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Tandem Mass Spectrometry , Transforming Growth Factor beta/chemistry , Transforming Growth Factor beta/genetics
4.
FEBS J ; 285(1): 101-114, 2018 01.
Article in English | MEDLINE | ID: mdl-29117645

ABSTRACT

TGFBIp is a constituent of the extracellular matrix in many human tissues including the cornea, where it is one of the most abundant proteins expressed. TGFBIp interacts with Type I, II, IV, VI, and XII collagens as well as several members of the integrin family, suggesting it plays an important role in maintaining structural integrity and possibly corneal transparency as well. Significantly, more than 60 point mutations within the TGFBI gene have been reported to result in aberrant TGFBIp folding and aggregation in the cornea, resulting in severe visual impairment and blindness. Several studies have focused on targeting TGFBIp in the cornea as a therapeutic approach to treat TGFBI-linked corneal dystrophies, but the effect of this approach on corneal homeostasis and matrix integrity remained unknown. In the current study, we evaluated the histological and proteomic profiles of corneas from TGFBI-deficient mice as well as potential redundant functions of the paralogous protein POSTN. The absence of TGFBIp in mouse corneas did not grossly affect the collagen scaffold, and POSTN is unable to compensate for loss of TGFBIp. Proteomic comparison of wild-type and TGFBI-/- mice revealed 11 proteins were differentially regulated, including Type VI and XII collagens. However, as these alterations did not manifest at the macroscopic and behavioral levels, these data support partial or complete TGFBI knockdown as a potential therapy against TGFBI-linked corneal dystrophies. Lastly, in situ hybridization verified TGFBI mRNA in the epithelial cells but not in other cell types, supportive of a therapy directed specifically at this lineage.


Subject(s)
Cornea/metabolism , Corneal Dystrophies, Hereditary/metabolism , Extracellular Matrix/metabolism , Proteomics/methods , Transforming Growth Factor beta/deficiency , Aged , Aged, 80 and over , Animals , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cornea/ultrastructure , Corneal Dystrophies, Hereditary/genetics , Corneal Dystrophies, Hereditary/therapy , Epithelial Cells/metabolism , Female , Gene Expression , Humans , Male , Mice , Mice, Knockout , Transforming Growth Factor beta/genetics
5.
Structure ; 25(11): 1740-1750.e2, 2017 11 07.
Article in English | MEDLINE | ID: mdl-28988748

ABSTRACT

A major cause of visual impairment, corneal dystrophies result from accumulation of protein deposits in the cornea. One of the proteins involved is transforming growth factor ß-induced protein (TGFBIp), an extracellular matrix component that interacts with integrins but also produces corneal deposits when mutated. Human TGFBIp is a multi-domain 683-residue protein, which contains one CROPT domain and four FAS1 domains. Its structure spans ∼120 Å and reveals that vicinal domains FAS1-1/FAS1-2 and FAS1-3/FAS1-4 tightly interact in an equivalent manner. The FAS1 domains are sandwiches of two orthogonal four-stranded ß sheets decorated with two three-helix insertions. The N-terminal FAS1 dimer forms a compact moiety with the structurally novel CROPT domain, which is a five-stranded all-ß cysteine-knot solely found in TGFBIp and periostin. The overall TGFBIp architecture discloses regions for integrin binding and that most dystrophic mutations cluster at both molecule ends, within domains FAS1-1 and FAS1-4.


Subject(s)
Extracellular Matrix Proteins/chemistry , Integrins/chemistry , Mutation , Protein Aggregates , Transforming Growth Factor beta/chemistry , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Corneal Dystrophies, Hereditary/genetics , Corneal Dystrophies, Hereditary/metabolism , Corneal Dystrophies, Hereditary/pathology , Crystallography, X-Ray , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , Humans , Integrins/genetics , Integrins/metabolism , Models, Molecular , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
6.
ACS Appl Mater Interfaces ; 8(24): 14975-9, 2016 Jun 22.
Article in English | MEDLINE | ID: mdl-27245371

ABSTRACT

Culturing cells on gradient nanopatterns provides a useful tool to explore cellular adhesion to mimics of the extracellular matrix or screen for cellular responses to nanopatterns. A method is presented to fabricate complex gradient protein patterns based on hole-mask colloidal lithography, which can generate nanopatterns in multiple materials and of multiple shapes. Gradients of gold structures were functionalized to form gradients of protein nanopatterns of different shapes (bars, dot pairs, and rings), where a key parameter was systematically varied in each gradient. Cells were grown on vitronectin nanopatterns, showing differential adhesion (spread area/focal adhesion size) along the gradients.


Subject(s)
Cell Adhesion , Cell Culture Techniques/methods , Extracellular Matrix/metabolism , Focal Adhesions , Nanotubes, Carbon , Vitronectin/metabolism
7.
Adv Mater ; 28(7): 1472-6, 2016 Feb 17.
Article in English | MEDLINE | ID: mdl-26650176

ABSTRACT

A novel combinatorial biomolecular nanopatterning method is reported, in which multiple biomolecular ligands can be patterned in multiple nanoscale dimensions on a single surface. The applicability of the combinatorial platform toward cell-biology applications is demonstrated by screening the adhesion behavior of a population of human dental pulp stem cell (hDPSC) on 64 combinations of nanopatterned extracellular matrix (ECM) proteins in parallel.


Subject(s)
Cell Culture Techniques/methods , Nanotechnology/methods , Stem Cells/cytology , Cell Adhesion , Dental Pulp/cytology , Humans
8.
Biomol NMR Assign ; 10(1): 25-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26275916

ABSTRACT

The transforming growth factor beta induced protein (TGFBIp) is a major protein component of the human cornea. Mutations occurring in TGFBIp may cause corneal dystrophies, which ultimately lead to loss of vision. The majority of the disease-causing mutations are located in the C-terminal domain of TGFBIp, referred as the fourth fascilin-1 (FAS1-4) domain. In the present study the FAS1-4 Ala546Thr, a mutation that causes lattice corneal dystrophy, was investigated in dimethylsulfoxide using liquid-state NMR spectroscopy, to enable H/D exchange strategies for identification of the core formed in mature fibrils. Isotope-labeled fibrillated FAS1-4 A546T was dissolved in a ternary mixture 95/4/1 v/v/v% dimethylsulfoxide/water/trifluoroacetic acid, to obtain and assign a reference 2D (1)H-(15)N HSQC spectrum for the H/D exchange analysis. Here, we report the near-complete assignments of backbone and aliphatic side chain (1)H, (13)C and (15)N resonances for unfolded FAS1-4 A546T at 25 °C.


Subject(s)
Dimethyl Sulfoxide/pharmacology , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/metabolism , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Nuclear Magnetic Resonance, Biomolecular , Protein Denaturation/drug effects , Transforming Growth Factor beta/chemistry , Transforming Growth Factor beta/metabolism , Amino Acid Sequence , Carbon Isotopes , Humans , Nitrogen Isotopes , Protein Domains , Tritium
9.
Biochemistry ; 54(36): 5546-56, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26305369

ABSTRACT

The human transforming growth factor ß-induced protein (TGFBIp) is involved in several types of corneal dystrophies where protein aggregation and amyloid fibril formation severely impair vision. Most disease-causing mutations are located in the last of four homologous fasciclin-1 (FAS1) domains of the protein, and it has been shown that when isolated, the fourth FAS1 domain (FAS1-4) mimics the behavior of full-length TGFBIp. In this study, we use molecular dynamics simulations and principal component analysis to study the wild-type FAS1-4 domain along with three disease-causing mutations (R555W, R555Q, and A546T) to decipher any internal difference in dynamical properties of the domains that may explain their varied stabilities and aggregation properties. In addition, we use a protein-protein docking method in combination with chemical cross-linking experiments and mass spectrometry of the cross-linked species to obtain information about interaction faces between identical FAS1-4 domains. The results show that the pathogenic mutations A546T and R555W affect the packing in the hydrophobic core of FAS1-4 in different directions. We further show that the FAS1-4 monomers associate using their ß-rich regions, consistent with peptides observed to be part of the amyloid fibril core in lattice corneal dystrophy patients.


Subject(s)
Amyloid/chemistry , Corneal Dystrophies, Hereditary/genetics , Extracellular Matrix Proteins/chemistry , Transforming Growth Factor beta/chemistry , Amyloid/genetics , Chromatography, Liquid , Computer Simulation , Cross-Linking Reagents/chemistry , Extracellular Matrix Proteins/genetics , Hydrophobic and Hydrophilic Interactions , Molecular Dynamics Simulation , Mutation , Succinimides/chemistry , Tandem Mass Spectrometry , Transforming Growth Factor beta/genetics
10.
Biochemistry ; 54(19): 2943-56, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25910219

ABSTRACT

Mutations in the transforming growth factor beta-induced (TGFBI) gene result in a group of hereditary diseases of the cornea that are collectively known as TGFBI corneal dystrophies. These mutations translate into amino acid substitutions mainly within the fourth fasciclin 1 domain (FAS1-4) of the transforming growth factor beta-induced protein (TGFBIp) and cause either amyloid or nonamyloid protein aggregates in the anterior and central parts of the cornea, depending on the mutation. The A546T substitution in TGFBIp causes lattice corneal dystrophy (LCD), which manifests as amyloid-type aggregates in the corneal stroma. We previously showed that the A546T substitution renders TGFBIp and the FAS1-4 domain thermodynamically less stable compared with the wild-type (WT) protein, and the mutant FAS1-4 is prone to amyloid formation in vitro. In the present study, we identified the core of A546T FAS1-4 amyloid fibrils. Significantly, we identified the Y571-R588 region of TGFBIp, which we previously found to be enriched in amyloid deposits in LCD patients. We further found that the Y571-R588 peptide seeded fibrillation of A546T FAS1-4, and, more importantly, we demonstrated that native TGFBIp aggregates in the presence of fibrils formed by the core peptide. Collectively, these data suggest an involvement of the Y571-R588 peptide in LCD pathophysiology.


Subject(s)
Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/metabolism , Transforming Growth Factor beta/chemistry , Transforming Growth Factor beta/metabolism , Corneal Dystrophies, Hereditary/metabolism , Corneal Stroma/metabolism , Humans , Microscopy, Electron, Transmission , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
11.
J Proteome Res ; 13(11): 4659-67, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-24846694

ABSTRACT

Fuchs' endothelial corneal dystrophy (FECD) is a major corneal disorder affecting the innermost part of the cornea, leading to visual impairment. As the morphological changes in FECD are mainly observed in the extracellular matrix of the Descemet's membrane/endothelial layer, we determined the protein profiles of diseased and control tissues using two relative quantitation MS methods. The first quantitation method, based on the areas of the extracted ion chromatograms, quantified the 51 and 48 most abundant proteins of the Descemet's membrane/endothelial layer in patient and control tissues, respectively, of which 10 were significantly regulated. The results indicated that the level of type VIII collagen was unaltered even though the protein previously has been shown to be implicated in familial early-onset forms of the disease. Using the second relative quantitation method, iTRAQ, we identified 22 differentially regulated proteins, many of which are extracellular proteins known to be involved in proper assembly of the basement membrane in other tissues. In total, 26 differentially regulated proteins were identified, of which 6 proteins were regulated in both methods. These results support that the morphological changes observed in FECD are caused in part by an aberrant assembly of the extracellular matrix within the Descemet's membrane/endothelial layer.


Subject(s)
Descemet Membrane/metabolism , Extracellular Matrix Proteins/metabolism , Fuchs' Endothelial Dystrophy/metabolism , Gene Expression Regulation/physiology , Proteomics/methods , Amino Acids/analysis , Chromatography, Liquid , Female , Humans , Male , Tandem Mass Spectrometry/methods
12.
J Biol Chem ; 289(9): 5462-6, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24398691

ABSTRACT

Non-healing wounds are a significant source of morbidity. This is particularly true for diabetic patients, who tend to develop chronic skin wounds. O-GlcNAc modification of serine and threonine residues is a common regulatory post-translational modification analogous to protein phosphorylation; increased intracellular protein O-GlcNAc modification has been observed in diabetic and hyperglycemic states. Two intracellular enzymes, UDP-N-acetylglucosamine-polypeptide ß-N-acetylglucosaminyl transferase (OGT) and O-GlcNAc-selective N-acetyl-ß-D-glucosaminidase (OGA), mediate addition and removal, respectively, of N-acetylglucosamine (GlcNAc) from intracellular protein substrates. Alterations in O-GlcNAc modification of intracellular proteins is linked to diabetes, and the increased levels of protein O-GlcNAc modification observed in diabetic tissues may in part explain some of the observed underlying pathophysiology that contributes to delayed wound healing. We have previously shown that increasing protein O-GlcNAc modification by overexpression of OGT in murine keratinocytes results in elevated protein O-GlcNAc modification and a hyperadhesive phenotype. This study was undertaken to explore the hypothesis that increased O-GlcNAc modification of cellular proteins in diabetic skin could contribute to the delayed wound healing observed in patients with diabetic skin ulcers. In the present study, we show that human keratinocytes cultured under hyperglycemic conditions display increased levels of O-GlcNAc modification as well as a delay in the rate of wound closure in vitro. We further show that specific knockdown of OGT by RNA interference (RNAi) reverses this effect, thereby opening up the opportunity for OGT-targeted therapies to promote wound healing in diabetic patients.


Subject(s)
Diabetes Complications/enzymology , Diabetes Complications/therapy , N-Acetylglucosaminyltransferases/metabolism , Skin/enzymology , Wound Healing , Wounds and Injuries/enzymology , Wounds and Injuries/therapy , Acetylglucosamine/genetics , Acetylglucosamine/metabolism , Animals , Cell Line , Diabetes Complications/genetics , Diabetes Complications/pathology , Gene Knockdown Techniques , Glycosylation , Humans , Keratinocytes/enzymology , Keratinocytes/pathology , Mice , N-Acetylglucosaminyltransferases/antagonists & inhibitors , N-Acetylglucosaminyltransferases/genetics , Protein Modification, Translational/genetics , RNA Interference , Skin/pathology , Wounds and Injuries/genetics , Wounds and Injuries/pathology
13.
Proteomics Clin Appl ; 8(3-4): 168-77, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24302499

ABSTRACT

PURPOSE: In this study, we investigated whether the phenotypic difference observed between two lattice corneal dystrophy type 1 (LCD type 1) cases caused by either a single A546D substitution or an A546D/P551Q double substitution in TGFBIp (transforming growth factor beta induced protein) can be ascribed to (i) a difference in the proteomes of corneal amyloid deposits, (ii) altered proteolysis of TGFBIp, or (iii) structural changes of TGFBIp introduced by the P551Q amino acid substitution. EXPERIMENTAL DESIGN: Amyloid deposits were isolated from the corneas of two siblings with LCD type 1 resulting from A546D/P551Q mutations in the TGFBI gene using laser capture microdissection and subsequently analyzed by LC-MS/MS. Proteolytic processing of TGFBIp was addressed by counting peptide spectra. Lastly, to study the possible effect of the P551Q substitution, recombinant FAS1-4 domain variants were subjected to in vitro stability assays. RESULTS: The amyloid proteomes and TGFBIp processing of the two A546D/P551Q LCD type 1 cases were similar to each other as well as to the A546D amyloid proteome previously reported by us. The stability assays revealed a minor destabilization of the FAS1-4 domain upon the addition of the P551Q mutation, moreover, it resulted in different accessibility to tryptic cleavage sites between the A546D and A546D/P551Q mutant FAS1-4 domain variants. CONCLUSION AND CLINICAL RELEVANCE: The difference in A546D and A546D/P551Q LCD type 1 phenotypes cannot be ascribed to altered corneal amyloid composition or altered in vivo proteolytic processing of TGFBIp. Instead, a small difference in thermodynamic stability introduced by the P551Q mutation most likely causes structural changes of TGFBIp. The MS proteomics data have been deposited to the ProteomeXchange with identifier PXD000307 (http://proteomecentral.proteomexchange.org/dataset/PXD000307).


Subject(s)
Cornea/pathology , Corneal Dystrophies, Hereditary/genetics , Extracellular Matrix Proteins/genetics , Transforming Growth Factor beta/genetics , Amino Acid Substitution/genetics , Amyloid/metabolism , Chromatography, Liquid , Cornea/metabolism , Corneal Dystrophies, Hereditary/metabolism , Corneal Dystrophies, Hereditary/pathology , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/chemistry , Humans , Laser Capture Microdissection , Mutation , Proteolysis , Tandem Mass Spectrometry , Transforming Growth Factor beta/chemistry
14.
Biochim Biophys Acta ; 1834(12): 2812-22, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24129074

ABSTRACT

Hereditary mutations in the transforming growth factor beta induced (TGFBI) gene cause phenotypically distinct corneal dystrophies characterized by protein deposition in cornea. We show here that the Arg555Trp mutant of the fourth fasciclin 1 (FAS1-4) domain of the protein (TGFBIp/keratoepithelin/ßig-h3), associated with granular corneal dystrophy type 1, is significantly less susceptible to proteolysis by thermolysin and trypsin than the WT domain. High-resolution liquid-state NMR of the WT and Arg555Trp mutant FAS1-4 domains revealed very similar structures except for the region around position 555. The Arg555Trp substitution causes Trp555 to be buried in an otherwise empty hydrophobic cavity of the FAS1-4 domain. The first thermolysin cleavage in the core of the FAS1-4 domain occurs on the N-terminal side of Leu558 adjacent to the Arg555 mutation. MD simulations indicated that the C-terminal end of helix α3' containing this cleavage site is less flexible in the mutant domain, explaining the observed proteolytic resistance. This structural change also alters the electrostatic properties, which may explain increased propensity of the mutant to aggregate in vitro with 2,2,2-trifluoroethanol. Based on our results we propose that the Arg555Trp mutation disrupts the normal degradation/turnover of corneal TGFBIp, leading to accumulation and increased propensity to aggregate through electrostatic interactions.


Subject(s)
Amino Acid Substitution , Corneal Dystrophies, Hereditary , Extracellular Matrix Proteins/chemistry , Mutation, Missense , Proteolysis , Transforming Growth Factor beta/chemistry , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Humans , Protein Stability , Protein Structure, Secondary , Protein Structure, Tertiary , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
15.
Biochemistry ; 52(16): 2821-7, 2013 Apr 23.
Article in English | MEDLINE | ID: mdl-23556985

ABSTRACT

TGFBIp, also known as keratoepithelin and ßig-h3, is among the most abundant proteins in the human cornea, and approximately 60% is associated with the insoluble fraction following extraction in sodium dodecyl sulfate (SDS) sample buffer. TGFBIp is of particular interest because a wide range of mutations causes amyloid or fuchsinophilic crystalloid deposits in the cornea leading to visual impairment. We show that the SDS-insoluble fraction of TGFBIp from porcine and human corneas is covalently linked via a reducible bond to the NC3 domain of type XII collagen in a TGFBIp:type XII collagen stoichiometric ratio of 2:1. Because type XII collagen is anchored to striated collagen fibers of the extracellular matrix, its interaction with TGFBIp is likely to provide anchoring for cells to the extracellular matrix through the integrin binding capability of TGFBIp. Furthermore, the TGFBIp-type XII collagen molecule will affect our understanding of the molecular pathogenesis of the TGFBI-linked corneal dystrophies.


Subject(s)
Collagen Type XII/chemistry , Collagen Type XII/metabolism , Cornea/chemistry , Extracellular Matrix Proteins/chemistry , Transforming Growth Factor beta/chemistry , Animals , Disulfides/chemistry , Disulfides/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Humans , Molecular Weight , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Solubility , Swine , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
16.
Mol Vis ; 19: 861-76, 2013.
Article in English | MEDLINE | ID: mdl-23592924

ABSTRACT

PURPOSE: Specific mutations in the transforming growth factor beta induced (TGFBI) gene are associated with lattice corneal dystrophy (LCD) type 1 and its variants. In this study, we performed an in-depth proteomic analysis of human corneal amyloid deposits associated with the heterozygous A546D mutation in TGFBI. METHODS: Corneal amyloid deposits and the surrounding corneal stroma were procured by laser capture microdissection from a patient with an A546D mutation in TGFBI. Proteins in the captured corneal samples and healthy corneal stroma were identified with liquid chromatography-tandem mass spectrometry and quantified by calculating exponentially modified Protein Abundance Index values. Mass spectrometry data were further compared for identifying enriched regions of transforming growth factor beta induced protein (TGFBIp/keratoepithelin/ßig-h3) and detecting proteolytic cleavage sites in TGFBIp. RESULTS: A C-terminal fragment of TGFBIp containing residues Y571-R588 derived from the fourth fasciclin 1 domain (FAS1-4), serum amyloid P-component, apolipoprotein A-IV, clusterin, and serine protease HtrA1 were significantly enriched in the amyloid deposits compared to the healthy cornea. The proteolytic cleavage sites in TGFBIp from the diseased cornea are in accordance with the activity of serine protease HtrA1. We also identified small amounts of the serine protease kallikrein-14 in the amyloid deposits. CONCLUSIONS: Corneal amyloid caused by the A546D mutation in TGFBI involves several proteins associated with other varieties of amyloidosis. The proteomic data suggest that the sequence 571-YHIGDEILVSGGIGALVR-588 contains the amyloid core of the FAS1-4 domain of TGFBIp and point at serine protease HtrA1 as the most likely candidate responsible for the proteolytic processing of amyloidogenic and aggregated TGFBIp, which explains the accumulation of HtrA1 in the amyloid deposits. With relevance to identifying serine proteases, we also found glia-derived nexin (protease-nexin 1) in the amyloid deposits, making this serine protease inhibitor a good candidate for the physiologically relevant inhibitor of one of the amyloid-associated serine proteases in the cornea and probably in other tissues. Noteworthy, the present results are in accordance with our findings from a previous study of corneal amyloid deposits caused by the V624M mutation in TGFBI, suggesting a common mechanism for lattice corneal dystrophies (LCDs) associated with mutations in the TGFBIp FAS1-4 domain.


Subject(s)
Cornea/metabolism , Cornea/pathology , Extracellular Matrix Proteins/metabolism , Plaque, Amyloid/metabolism , Serine Endopeptidases/metabolism , Transforming Growth Factor beta/metabolism , Aged , Amino Acid Sequence , Cluster Analysis , Corneal Dystrophies, Hereditary/metabolism , Corneal Dystrophies, Hereditary/pathology , Corneal Stroma/metabolism , Corneal Stroma/pathology , Extracellular Matrix Proteins/chemistry , Female , High-Temperature Requirement A Serine Peptidase 1 , Humans , Microdissection , Molecular Sequence Data , Protease Inhibitors/metabolism , Protein Structure, Tertiary , Proteolysis , Proteomics , Sequence Alignment , Tandem Mass Spectrometry , Transforming Growth Factor beta/chemistry , Trypsin/metabolism
17.
J Biol Chem ; 287(41): 34730-42, 2012 Oct 05.
Article in English | MEDLINE | ID: mdl-22893702

ABSTRACT

Mutations in the transforming growth factor ß-induced protein (TGFBIp) are linked to the development of corneal dystrophies in which abnormal protein deposition in the cornea leads to a loss of corneal transparency and ultimately blindness. Different mutations give rise to phenotypically distinct corneal dystrophies. Most mutations are located in the fourth fasciclin-1 domain (FAS1-4). The amino acid substitution A546T in the FAS1-4 domain is linked to the development of lattice corneal dystrophy with amyloid deposits in the superficial and deep stroma, classifying it as an amyloid disease. Here we provide a detailed description of the fibrillation of the isolated FAS1-4 domain carrying the A546T substitution. The A546T substitution leads to a significant destabilization of FAS1-4 and induces a partially folded structure with increased surface exposure of hydrophobic patches. The mutation also leads to two distinct fibril morphologies. Long straight fibrils composed of pure ß-sheet structure are formed at lower concentrations, whereas short and curly fibrils containing a mixture of α-helical and ß-sheet structures are formed at higher concentrations. The formation of short and curly fibrils is preceded by the formation of a small number of oligomeric species with high membrane permeabilization potential and rapid fibril formation. The long straight fibrils are formed more slowly and through progressively bigger oligomers that lose their membrane permeabilization potential as fibrillation proceeds beyond the lag phase. These different fibril classes and associated biochemical differences may lead to different clinical symptoms associated with the mutation.


Subject(s)
Amyloid/chemistry , Extracellular Matrix Proteins/chemistry , Protein Multimerization , Transforming Growth Factor beta/chemistry , Amino Acid Substitution , Amyloid/genetics , Amyloid/metabolism , Cell Membrane/genetics , Cell Membrane/metabolism , Cornea/metabolism , Cornea/pathology , Corneal Dystrophies, Hereditary/genetics , Corneal Dystrophies, Hereditary/metabolism , Corneal Dystrophies, Hereditary/pathology , Extracellular Matrix Proteins/metabolism , Mutation, Missense , Permeability , Protein Structure, Quaternary , Protein Structure, Secondary , Transforming Growth Factor beta/metabolism
18.
Exp Eye Res ; 96(1): 163-70, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22155582

ABSTRACT

Different types of granular corneal dystrophy (GCD) and lattice corneal dystrophy (LCD) are associated with mutations in the transforming growth factor beta induced gene (TGFBI). These dystrophies are characterized by the formation of non-amyloid granular deposits (GCDs) and amyloid (LCD type 1 and its variants) in the cornea. Typical corneal non-amyloid deposits from GCD type 2 (R124H), amyloid from a variant of LCD type 1 (V624M) and disease-free tissue controls were procured by laser capture microdissection and analyzed by tandem mass spectrometry. Label-free quantitative comparisons of deposits and controls suggested that the non-amyloid sample (R124H) specifically accumulated transforming growth factor beta induced protein (TGFBIp/keratoepithelin/ßig-h3), serum amyloid P-component, clusterin, type III collagen, keratin 3, and histone H3-like protein. The amyloid (V624M) similarly accumulated serum amyloid P-component and clusterin but also a C-terminal fragment of TGFBIp containing residues Y571-R588 derived from the fourth fasciclin-1 domain (FAS1-4), apolipoprotein E and apolipoprotein A-IV. Significantly, analyses of the amyloid sample also revealed the presence of the serine protease Htr (High-temperature requirement) A1 and a number of proteolytic cleavage sites in the FAS1-4 domain of TGFBIp. These cleavage sites were consistent with the ligand binding and proteolytic activity of HtrA1 suggesting that it plays a role in the proteolytic processing of the amyloidogenic FAS1-4 domain. Taken together, the data suggest that the amyloidogenic-prone region of the fourth FAS1 domain of TGFBIp encompasses the Y571-R588 peptide and that HtrA1 is involved in the proteolytic processing of TGFBIp-derived amyloid in vivo.


Subject(s)
Amyloidosis, Familial/metabolism , Corneal Dystrophies, Hereditary/metabolism , Corneal Stroma/metabolism , Extracellular Matrix Proteins/genetics , Mutation , Plaque, Amyloid/metabolism , Transforming Growth Factor beta/genetics , Amyloidosis, Familial/genetics , Apolipoproteins/metabolism , Chromatography, Liquid , Clusterin/metabolism , Collagen Type III/metabolism , Corneal Dystrophies, Hereditary/genetics , Humans , Keratin-3/metabolism , Laser Capture Microdissection , Proteolysis , Proteomics , Serum Amyloid P-Component/metabolism , Tandem Mass Spectrometry
19.
J Biol Chem ; 286(7): 4951-8, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21135107

ABSTRACT

Mutations in the human TGFBI gene encoding TGFBIp have been linked to protein deposits in the cornea leading to visual impairment. The protein consists of an N-terminal Cys-rich EMI domain and four consecutive fasciclin 1 (FAS1) domains. We have compared the stabilities of wild-type (WT) human TGFBIp and six mutants known to produce phenotypically distinct deposits in the cornea. Amino acid substitutions in the first FAS1 (FAS1-1) domain (R124H, R124L, and R124C) did not alter the stability. However, substitutions within the fourth FAS1 (FAS1-4) domain (A546T, R555Q, and R555W) affected the overall stability of intact TGFBIp revealing the following stability ranking R555W>WT>R555Q>A546T. Significantly, the stability ranking of the isolated FAS1-4 domains mirrored the behavior of the intact protein. In addition, it was linked to the aggregation propensity as the least stable mutant (A546T) forms amyloid fibrils while the more stable variants generate non-amyloid amorphous deposits in vivo. Significantly, the data suggested that both an increase and a decrease in the stability of FAS1-4 may unleash a disease mechanism. In contrast, amino acid substitutions in FAS1-1 did not affect the stability of the intact TGFBIp suggesting that molecular the mechanism of disease differs depending on the FAS1 domain carrying the mutation.


Subject(s)
Amino Acid Substitution , Amyloid/metabolism , Cornea/metabolism , Corneal Dystrophies, Hereditary/metabolism , Extracellular Matrix Proteins/metabolism , Mutation, Missense , Transforming Growth Factor beta/metabolism , Amyloid/genetics , Corneal Dystrophies, Hereditary/genetics , Extracellular Matrix Proteins/genetics , HEK293 Cells , Humans , Protein Stability , Protein Structure, Tertiary , Transforming Growth Factor beta/genetics
20.
Exp Eye Res ; 90(1): 57-62, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19788893

ABSTRACT

Transforming growth factor beta induced protein (TGFBIp, also named keratoepithelin) is an extracellular matrix protein abundant in the cornea. The purpose of this study was to determine the expression and processing of TGFBIp in the normal human cornea during postnatal development and aging. TGFBIp in corneas from individuals ranging from six months to 86 years of age was detected and quantified by immunoblotting. The level of TGFBIp in the cornea increases about 30% between 6 and 14 years of age, and adult corneas contain 0.7-0.8 microg TGFBIp per mg wet tissue. Two-dimensional (2-D) immunoblots of the corneal extracts showed a characteristic "zig-zag" pattern formed by different lower-molecular mass TGFBIp isoforms (30-60 kDa). However, the relative abundance of the different isoforms was different between infant corneas (<1 year) and the child/adult corneas (>6 years). Matrix-assisted laser desorption/ionization-time-of-flight mass spectrometry (MALDI-TOF MS) data of TGFBIp isoforms separated on large 2-D gels show that TGFBIp is proteolytically processed from the N-terminus. This observation was supported by in silico 2-D gel electrophoresis showing that sequential proteolytical trimming events from the N-terminus of mature TGFBIp generate TGFBIp isoforms which form a similar "zig-zag" pattern when separated by 2-D polyacrylamide gel electrophoresis (PAGE). This study shows that in humans TGFBIp is more abundant in mature corneas than in the developing cornea and that the processing of TGFBIp changes during postnatal development of the cornea. In addition, TGFBIp appears to be degraded in a highly orchestrated manner in the normal human cornea with the resulting C-terminal fragments being retained in the cornea. The age-related changes in the expression and processing of corneal TGFBIp suggests that TGFBIp may play a role in the postnatal development and maturation of the cornea. Furthermore, these observations may be relevant to the age at which mutant TGFBIp deposits in the cornea in those dystrophies caused by mutations in the transforming growth factor beta induced gene (TGFBI) as well as the mechanisms of corneal protein deposition.


Subject(s)
Aging/physiology , Cornea/growth & development , Cornea/metabolism , Extracellular Matrix Proteins/metabolism , Transforming Growth Factor beta/metabolism , Adolescent , Aged, 80 and over , Child , Electrophoresis, Gel, Two-Dimensional , Female , Humans , Immunoblotting , Infant , Male , Middle Aged , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...