Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
Add more filters

Publication year range
1.
J Biol Chem ; 299(12): 105459, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37977222

ABSTRACT

The collagen IVα345 (Col-IVα345) scaffold, the major constituent of the glomerular basement membrane (GBM), is a critical component of the kidney glomerular filtration barrier. In Alport syndrome, affecting millions of people worldwide, over two thousand genetic variants occur in the COL4A3, COL4A4, and COL4A5 genes that encode the Col-IVα345 scaffold. Variants cause loss of scaffold, a suprastructure that tethers macromolecules, from the GBM or assembly of a defective scaffold, causing hematuria in nearly all cases, proteinuria, and often progressive kidney failure. How these variants cause proteinuria remains an enigma. In a companion paper, we found that the evolutionary emergence of the COL4A3, COL4A4, COL4A5, and COL4A6 genes coincided with kidney emergence in hagfish and shark and that the COL4A3 and COL4A4 were lost in amphibians. These findings opened an experimental window to gain insights into functionality of the Col-IVα345 scaffold. Here, using tissue staining, biochemical analysis and TEM, we characterized the scaffold chain arrangements and the morphology of the GBM of hagfish, shark, frog, and salamander. We found that α4 and α5 chains in shark GBM and α1 and α5 chains in amphibian GBM are spatially separated. Scaffolds are distinct from one another and from the mammalian Col-IVα345 scaffold, and the GBM morphologies are distinct. Our findings revealed that the evolutionary emergence of the Col-IVα345 scaffold enabled the genesis of a compact GBM that functions as an ultrafilter. Findings shed light on the conundrum, defined decades ago, whether the GBM or slit diaphragm is the primary filter.


Subject(s)
Collagen Type IV , Glomerular Basement Membrane , Mammals , Animals , Anura , Collagen Type IV/classification , Collagen Type IV/genetics , Collagen Type IV/metabolism , Glomerular Basement Membrane/chemistry , Glomerular Basement Membrane/metabolism , Glomerular Basement Membrane/physiology , Hagfishes , Mammals/genetics , Mammals/metabolism , Mammals/physiology , Sharks , Species Specificity , Urodela
2.
PLoS Genet ; 17(2): e1009339, 2021 02.
Article in English | MEDLINE | ID: mdl-33524049

ABSTRACT

Heat shock protein 47 (HSP47), encoded by the SERPINH1 gene, is a molecular chaperone essential for correct folding of collagens. We report a homozygous p.(R222S) substitution in HSP47 in a child with severe osteogenesis imperfecta leading to early demise. p.R222 is a highly conserved residue located within the collagen interacting surface of HSP47. Binding assays show a significantly reduced affinity of HSP47-R222S for type I collagen. This altered interaction leads to posttranslational overmodification of type I procollagen produced by dermal fibroblasts, with increased glycosylation and/or hydroxylation of lysine and proline residues as shown by mass spectrometry. Since we also observed a normal intracellular folding and secretion rate of type I procollagen, this overmodification cannot be explained by prolonged exposure of the procollagen molecules to the modifying hydroxyl- and glycosyltransferases, as is commonly observed in other types of OI. We found significant upregulation of several molecular chaperones and enzymes involved in procollagen modification and folding on Western blot and RT-qPCR. In addition, we showed that an imbalance in binding of HSP47-R222S to unfolded type I collagen chains in a gelatin sepharose pulldown assay results in increased binding of other chaperones and modifying enzymes. The elevated expression and binding of this molecular ensemble to type I procollagen suggests a compensatory mechanism for the aberrant binding of HSP47-R222S, eventually leading to overmodification of type I procollagen chains. Together, these results illustrate the importance of HSP47 for proper posttranslational modification and provide insights into the molecular pathomechanisms of the p.(R222S) alteration in HSP47, which leads to a severe OI phenotype.


Subject(s)
Collagen Type I/genetics , HSP47 Heat-Shock Proteins/genetics , Mutation, Missense , Osteogenesis Imperfecta/genetics , Amino Acid Sequence , Cells, Cultured , Child, Preschool , Collagen Type I/metabolism , Fatal Outcome , Female , HSP47 Heat-Shock Proteins/chemistry , HSP47 Heat-Shock Proteins/metabolism , Humans , Infant , Infant, Newborn , Models, Molecular , Osteogenesis Imperfecta/metabolism , Protein Binding , Protein Domains , Protein Processing, Post-Translational , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid
3.
J Biol Chem ; 296: 100453, 2021.
Article in English | MEDLINE | ID: mdl-33631195

ABSTRACT

Collagen is the most abundant protein in humans. It has a characteristic triple-helix structure and is heavily posttranslationally modified. The complex biosynthesis of collagen involves processing by many enzymes and chaperones in the rough endoplasmic reticulum. Lysyl hydroxylase 1 (LH1) is required to hydroxylate lysine for cross-linking and carbohydrate attachment within collagen triple helical sequences. Additionally, a recent study of prolyl 3-hydroxylase 3 (P3H3) demonstrated that this enzyme may be critical for LH1 activity; however, the details surrounding its involvement remain unclear. If P3H3 is an LH1 chaperone that is critical for LH1 activity, P3H3 and LH1 null mice should display a similar deficiency in lysyl hydroxylation. To test this hypothesis, we compared the amount and location of hydroxylysine in the triple helical domains of type V and I collagen from P3H3 null, LH1 null, and wild-type mice. The amount of hydroxylysine in type V collagen was reduced in P3H3 null mice, but surprisingly type V collagen from LH1 null mice contained as much hydroxylysine as type V collagen from wild-type mice. In type I collagen, our results indicate that LH1 plays a global enzymatic role in lysyl hydroxylation. P3H3 is also involved in lysyl hydroxylation, particularly at cross-link formation sites, but is not required for all lysyl hydroxylation sites. In summary, our study suggests that LH1 and P3H3 likely have two distinct mechanisms to recognize different collagen types and to distinguish cross-link formation sites from other sites in type I collagen.


Subject(s)
Collagen Type I/metabolism , Collagen Type V/metabolism , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/metabolism , Procollagen-Proline Dioxygenase/metabolism , Animals , Collagen/genetics , Collagen/metabolism , Collagen Type I/genetics , Collagen Type V/genetics , Endoplasmic Reticulum, Rough/metabolism , Hydroxylation , Hydroxylysine/metabolism , Lysine/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Procollagen-Proline Dioxygenase/genetics , Protein Conformation , Protein Processing, Post-Translational/genetics
4.
J Biol Chem ; 293(16): 5987-5999, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29491144

ABSTRACT

Type IV collagen is a major component of the basement membrane and interacts with numerous other basement membrane proteins. Many of these interactions are poorly characterized. Type IV collagen is abundantly post-translationally modified with 3-hydroxyproline (3-Hyp), but 3-Hyp's biochemical role in type IV collagen's interactions with other proteins is not well established. In this work, we present binding data consistent with a major role of 3-Hyp in interactions of collagen IV with glycoprotein VI and nidogens 1 and 2. The increased binding interaction between type IV collagen without 3-Hyp and glycoprotein VI has been the subject of some controversy, which we sought to explore, whereas the lack of binding of nidogens to type IV collagen without 3-Hyp is novel. Using tandem MS, we show that the putative glycoprotein VI-binding site is 3-Hyp-modified in WT PFHR-9 type IV collagen, but not in PFHR-9 cells in which prolyl-3-hydroxylase 2 (P3H2) has been knocked out (KO). Moreover, we observed altered 3-Hyp occupancy across many other sites. Using amino acid analysis of type IV collagen from the WT and P3H2 KO cell lines, we confirm that P3H2 is the major, but not the only 3-Hyp-modifying enzyme of type IV collagen. These findings underscore the importance of post-translational modifications of type IV collagen for interactions with other proteins.


Subject(s)
Collagen Type IV/metabolism , Hydroxyproline/metabolism , Membrane Glycoproteins/metabolism , Platelet Membrane Glycoproteins/metabolism , Protein Interaction Maps , Animals , Calcium-Binding Proteins , Cell Adhesion Molecules , Cell Line , Mice , Protein Processing, Post-Translational
5.
J Biol Chem ; 293(35): 13707-13716, 2018 08 31.
Article in English | MEDLINE | ID: mdl-30002123

ABSTRACT

The build-up of diversified and tissue-specific assemblies of extracellular matrix (ECM) proteins depends on secreted and cell surface-located molecular arrays that coordinate ECM proteins into discrete designs. The family of small leucine-rich proteins (SLRPs) associates with and dictates the structure of fibrillar collagens, which form the backbone of most ECM types. However, whether SLRPs form complexes with proteins other than collagens is unclear. Here, we demonstrate that heat shock protein 47 (Hsp47), a well-established endoplasmic reticulum-resident collagen chaperone, also binds the SLRPs decorin, lumican, and fibromodulin with affinities comparable with that in the Hsp47-type I collagen interaction. Furthermore, we show that a lack of Hsp47 inhibits the cellular secretion of decorin and lumican. Our results expand the understanding of the concerted molecular interactions that control the secretion and organization of a functional collagenous ECM.


Subject(s)
Collagen Type I/metabolism , Decorin/metabolism , Fibromodulin/metabolism , HSP47 Heat-Shock Proteins/metabolism , Lumican/metabolism , Protein Interaction Maps , Animals , Cell Line , Endoplasmic Reticulum/metabolism , Humans , Mice , NIH 3T3 Cells
6.
Proc Natl Acad Sci U S A ; 113(41): E6036-E6044, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27679847

ABSTRACT

Extracellular matrix (ECM) proteins are biosynthesized in the rough endoplasmic reticulum (rER) and transported via the Golgi apparatus to the extracellular space. The coat protein complex II (COPII) transport vesicles are approximately 60-90 nm in diameter. However, several ECM molecules are much larger, up to several hundreds of nanometers. Therefore, special COPII vesicles are required to coat and transport these molecules. Transmembrane Protein Transport and Golgi Organization 1 (TANGO1) facilitates loading of collagens into special vesicles. The Src homology 3 (SH3) domain of TANGO1 was proposed to recognize collagen molecules, but how the SH3 domain recognizes various types of collagen is not understood. Moreover, how are large noncollagenous ECM molecules transported from the rER to the Golgi? Here we identify heat shock protein (Hsp) 47 as a guide molecule directing collagens to special vesicles by interacting with the SH3 domain of TANGO1. We also consider whether the collagen secretory model applies to other large ECM molecules.


Subject(s)
Extracellular Matrix Proteins/metabolism , Aryl Hydrocarbon Receptor Nuclear Translocator/chemistry , Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , COP-Coated Vesicles , Collagen/metabolism , Endoplasmic Reticulum/metabolism , Extracellular Matrix , Fibrillin-1/metabolism , Gene Expression , Golgi Apparatus/metabolism , HSP47 Heat-Shock Proteins/metabolism , Humans , Intracellular Space/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Transport , Recombinant Proteins , src Homology Domains/genetics
7.
J Biol Chem ; 292(42): 17216-17224, 2017 10 20.
Article in English | MEDLINE | ID: mdl-28860186

ABSTRACT

Collagen is the most abundant protein in the extracellular matrix in humans and is critical to the integrity and function of many musculoskeletal tissues. A molecular ensemble comprising more than 20 molecules is involved in collagen biosynthesis in the rough endoplasmic reticulum. Two proteins, heat shock protein 47 (Hsp47/SERPINH1) and 65-kDa FK506-binding protein (FKBP65/FKBP10), have been shown to play important roles in this ensemble. In humans, autosomal recessive mutations in both genes cause similar osteogenesis imperfecta phenotypes. Whereas it has been proposed that Hsp47 and FKBP65 interact in the rough endoplasmic reticulum, there is neither clear evidence for this interaction nor any data regarding their binding affinities for each other. In this study using purified endogenous proteins, we examined the interaction between Hsp47, FKBP65, and collagen and also determined their binding affinities and functions in vitro Hsp47 and FKBP65 show a direct but weak interaction, and FKBP65 prefers to interact with Hsp47 rather than type I collagen. Our results suggest that a weak interaction between Hsp47 and FKBP65 confers mutual molecular stability and also allows for a synergistic effect during collagen folding. We also propose that Hsp47 likely acts as a hub molecule during collagen folding and secretion by directing other molecules to reach their target sites on collagens. Our findings may explain why osteogenesis imperfecta-causing mutations in both genes result in similar phenotypes.


Subject(s)
Avian Proteins/chemistry , Collagen/chemistry , Endoplasmic Reticulum/chemistry , HSP47 Heat-Shock Proteins/chemistry , Protein Folding , Tacrolimus Binding Proteins/chemistry , Animals , Avian Proteins/genetics , Avian Proteins/metabolism , Chick Embryo , Chickens , Collagen/genetics , Collagen/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , HSP47 Heat-Shock Proteins/genetics , HSP47 Heat-Shock Proteins/metabolism , Humans , Mutation , Osteogenesis Imperfecta/genetics , Osteogenesis Imperfecta/metabolism , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism
8.
J Biol Chem ; 292(22): 9273-9282, 2017 06 02.
Article in English | MEDLINE | ID: mdl-28385890

ABSTRACT

Extracellular matrix proteins are biosynthesized in the rough endoplasmic reticulum (rER), and the triple-helical protein collagen is the most abundant extracellular matrix component in the human body. Many enzymes, molecular chaperones, and post-translational modifiers facilitate collagen biosynthesis. Collagen contains a large number of proline residues, so the cis/trans isomerization of proline peptide bonds is the rate-limiting step during triple-helix formation. Accordingly, the rER-resident peptidyl prolyl cis/trans isomerases (PPIases) play an important role in the zipper-like triple-helix formation in collagen. We previously described this process as "Ziploc-ing the structure" and now provide additional information on the activity of individual rER PPIases. We investigated the substrate preferences of these PPIases in vitro using type III collagen, the unhydroxylated quarter fragment of type III collagen, and synthetic peptides as substrates. We observed changes in activity of six rER-resident PPIases, cyclophilin B (encoded by the PPIB gene), FKBP13 (FKBP2), FKBP19 (FKBP11), FKBP22 (FKBP14), FKBP23 (FKBP7), and FKBP65 (FKBP10), due to posttranslational modifications of proline residues in the substrate. Cyclophilin B and FKBP13 exhibited much lower activity toward post-translationally modified substrates. In contrast, FKBP19, FKBP22, and FKBP65 showed increased activity toward hydroxyproline-containing peptide substrates. Moreover, FKBP22 showed a hydroxyproline-dependent effect by increasing the amount of refolded type III collagen in vitro and FKBP19 seems to interact with triple helical type I collagen. Therefore, we propose that hydroxyproline modulates the rate of Ziploc-ing of the triple helix of collagen in the rER.


Subject(s)
Collagen Type III/chemistry , Collagen Type I/chemistry , Cyclophilins/chemistry , Endoplasmic Reticulum/enzymology , Protein Processing, Post-Translational , Tacrolimus Binding Proteins/chemistry , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type III/metabolism , Cyclophilins/genetics , Cyclophilins/metabolism , Endoplasmic Reticulum/genetics , Humans , Hydroxyproline/chemistry , Hydroxyproline/metabolism , Protein Domains , Substrate Specificity , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism
9.
Am J Hum Genet ; 96(3): 425-31, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-25683117

ABSTRACT

Cole-Carpenter syndrome is a severe bone fragility disorder that is characterized by frequent fractures, craniosynostosis, ocular proptosis, hydrocephalus, and distinctive facial features. To identify the cause of Cole-Carpenter syndrome in the two individuals whose clinical results were presented in the original description of this disorder, we performed whole-exome sequencing of genomic DNA samples from both individuals. The two unrelated individuals had the same heterozygous missense mutation in exon 9 of P4HB (NM_000918.3: c.1178A>G [p.Tyr393Cys]), the gene that encodes protein disulfide isomerase (PDI). In one individual, the P4HB mutation had arisen de novo, whereas in the other the mutation was transmitted from the clinically unaffected father who was a mosaic carrier of the variant. The mutation was located in the C-terminal disulfide isomerase domain of PDI, sterically close to the enzymatic center, and affected disulfide isomerase activity in vitro. Skin fibroblasts showed signs of increased endoplasmic reticulum stress, but despite the reported importance of PDI for collagen type I production, the rate of collagen type I secretion appeared normal. In conclusion, Cole-Carpenter syndrome is caused by a specific de novo mutation in P4HB that impairs the disulfide isomerase activity of PDI.


Subject(s)
Craniosynostoses/genetics , Eye Abnormalities/genetics , Heterozygote , Hydrocephalus/genetics , Mutation, Missense , Osteogenesis Imperfecta/genetics , Procollagen-Proline Dioxygenase/genetics , Protein Disulfide-Isomerases/genetics , Child, Preschool , Female , Gene Frequency , Humans , Infant , Male , Pedigree , Procollagen-Proline Dioxygenase/metabolism , Protein Conformation , Protein Disulfide-Isomerases/metabolism , Protein Folding , Sequence Analysis, DNA
10.
Am J Hum Genet ; 96(6): 979-85, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26027498

ABSTRACT

Secreted protein, acidic, cysteine-rich (SPARC) is a glycoprotein that binds to collagen type I and other proteins in the extracellular matrix. Using whole-exome sequencing to identify the molecular defect in two unrelated girls with severe bone fragility and a clinical diagnosis of osteogenesis imperfecta type IV, we identified two homozygous variants in SPARC (GenBank: NM_003118.3; c.497G>A [p.Arg166His] in individual 1; c.787G>A [p.Glu263Lys] in individual 2). Published modeling and site-directed mutagenesis studies had previously shown that the residues substituted by these mutations form an intramolecular salt bridge in SPARC and are essential for the binding of SPARC to collagen type I. The amount of SPARC secreted by skin fibroblasts was reduced in individual 1 but appeared normal in individual 2. The migration of collagen type I alpha chains produced by these fibroblasts was mildly delayed on SDS-PAGE gel, suggesting some overmodification of collagen during triple helical formation. Pulse-chase experiments showed that collagen type I secretion was mildly delayed in skin fibroblasts from both individuals. Analysis of an iliac bone sample from individual 2 showed that trabecular bone was hypermineralized on the material level. In conclusion, these observations show that homozygous mutations in SPARC can give rise to severe bone fragility in humans.


Subject(s)
Models, Molecular , Mutation, Missense/genetics , Osteogenesis Imperfecta/genetics , Osteogenesis Imperfecta/pathology , Osteonectin/genetics , Amino Acid Sequence , Base Sequence , Collagen Type I/metabolism , Electrophoresis, Polyacrylamide Gel , Exome/genetics , Female , Genes, Recessive/genetics , Humans , Immunoblotting , Molecular Sequence Data , Mutagenesis, Site-Directed , Osteonectin/chemistry , Osteonectin/metabolism , Pedigree , Protein Conformation , Sequence Alignment , Sequence Analysis, DNA
11.
Am J Respir Cell Mol Biol ; 57(1): 77-90, 2017 07.
Article in English | MEDLINE | ID: mdl-28257580

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is characterized by excessive deposition of extracellular matrix, in particular, collagens. Two IPF therapeutics, nintedanib and pirfenidone, decelerate lung function decline, but their underlying mechanisms of action are poorly understood. In this study, we sought to analyze their effects on collagen synthesis and maturation at important regulatory levels. Primary human fibroblasts from patients with IPF and healthy donors were treated with nintedanib (0.01-1.0 µM) or pirfenidone (100-1,000 µM) in the absence or presence of transforming growth factor-ß1. Effects on collagen, fibronectin, FKBP10, and HSP47 expression, and collagen I and III secretion, were analyzed by quantitative polymerase chain reaction and Western blot. The appearance of collagen fibrils was monitored by scanning electron microscopy, and the kinetics of collagen fibril assembly was assessed using a light-scattering approach. In IPF fibroblasts, nintedanib reduced the expression of collagen I and V, fibronectin, and FKBP10 and attenuated the secretion of collagen I and III. Pirfenidone also down-regulated collagen V but otherwise showed fewer and less pronounced effects. By and large, the effects were similar in donor fibroblasts. For both drugs, electron microscopy of IPF fibroblast cultures revealed fewer and thinner collagen fibrils compared with untreated controls. Finally, both drugs dose-dependently delayed fibril formation of purified collagen I. In summary, both drugs act on important regulatory levels in collagen synthesis and processing. Nintedanib was more effective in down-regulating profibrotic gene expression and collagen secretion. Importantly, both drugs inhibited collagen I fibril formation and caused a reduction in and an altered appearance of collagen fibril bundles, representing a completely novel mechanism of action for both drugs.


Subject(s)
Fibrillar Collagens/metabolism , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/pathology , Indoles/therapeutic use , Pyridones/therapeutic use , Collagen Type I/metabolism , Down-Regulation/drug effects , Extracellular Matrix Proteins/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , HSP47 Heat-Shock Proteins/metabolism , Humans , Indoles/pharmacology , Lung/pathology , Protein Processing, Post-Translational/drug effects , Pyridones/pharmacology , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism , Transcription, Genetic/drug effects
12.
Proc Natl Acad Sci U S A ; 111(1): 161-6, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24368846

ABSTRACT

Collagens constitute nearly 30% of all proteins in our body. Type IV collagen is a major and crucial component of basement membranes. Collagen chains undergo several posttranslational modifications that are indispensable for proper collagen function. One of these modifications, prolyl 3-hydroxylation, is accomplished by a family of prolyl 3-hydroxylases (P3H1, P3H2, and P3H3). The present study shows that P3H2-null mice are embryonic-lethal by embryonic day 8.5. The mechanism of the unexpectedly early lethality involves the interaction of non-3-hydroxylated embryonic type IV collagen with the maternal platelet-specific glycoprotein VI (GPVI). This interaction results in maternal platelet aggregation, thrombosis of the maternal blood, and death of the embryo. The phenotype is completely rescued by producing double KOs of P3H2 and GPVI. Double nulls are viable and fertile. Under normal conditions, subendothelial collagens bear the GPVI-binding sites that initiate platelet aggregation upon blood exposure during injuries. In type IV collagen, these sites are normally 3-hydroxylated. Thus, prolyl 3-hydroxylation of type IV collagen has an important function preventing maternal platelet aggregation in response to the early developing embryo. A unique link between blood coagulation and the ECM is established. The newly described mechanism may elucidate some unexplained fetal losses in humans, where thrombosis is often observed at the maternal/fetal interface. Moreover, epigenetic silencing of P3H2 in breast cancers implies that the interaction between GPVI and non-3-hydroxylated type IV collagen might also play a role in the progression of malignant tumors and metastasis.


Subject(s)
Collagen Type IV/metabolism , Procollagen-Proline Dioxygenase/metabolism , Amino Acid Sequence , Animals , Blood Coagulation , Cattle , Collagen Type IV/chemistry , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Humans , Hydroxylation , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Phenotype , Platelet Aggregation , Procollagen-Proline Dioxygenase/chemistry , Protein Structure, Tertiary , Thrombosis , Time Factors
13.
Biochim Biophys Acta ; 1850(10): 1983-93, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25583561

ABSTRACT

BACKGROUND: Protein folding is crucial for proteins' specific functions and is facilitated by various types of enzymes and molecular chaperones. The peptidyl prolyl cis/trans isomerases (PPIase) are one of these families of enzymes. They ubiquitously exist inside the cell and there are eight PPIases in the rough endoplasmic reticulum (rER), a compartment where the folding of most secreted proteins occurs. SCOPE OF REVIEW: We review the functional and structural aspects of individual rER resident PPIases. Furthermore, we specifically discuss the role of these PPIases during collagen biosynthesis, since collagen is the most abundant protein in humans, is synthesized in the rER, and contains a proportionally high number of proline residues. MAJOR CONCLUSIONS: The rER resident PPIases recognize different sets of substrates and facilitate their folding. Although they are clearly catalysts for protein folding, they also have more broad and multifaceted functions. We propose that PPIases coordinate collagen biosynthesis in the rER. GENERAL SIGNIFICANCE: This review expands our understanding of collagen biosynthesis by explaining the influence of novel indirect mechanisms of regulating folding and this is also explored for PPIases. We also suggest future directions of research to obtain a better understanding of collagen biosynthesis and functions of PPIases in the rER. This article is part of a Special Issue entitled Proline-directed Foldases: Cell Signaling Catalysts and Drug Targets.


Subject(s)
Collagen/biosynthesis , Endoplasmic Reticulum/metabolism , Peptidylprolyl Isomerase/metabolism , Protein Folding , Animals , Humans , Protein Structure, Secondary
14.
J Biol Chem ; 289(26): 18189-201, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24821723

ABSTRACT

The biosynthesis of collagens occurs in the rough endoplasmic reticulum and requires a large numbers of molecular chaperones, foldases, and post-translational modification enzymes. Collagens contain a large number of proline residues that are post-translationally modified to 3-hydroxyproline or 4-hydroxyproline, and the rate-limiting step in formation of the triple helix is the cis-trans isomerization of peptidyl-proline bonds. This step is catalyzed by peptidyl-prolyl cis-trans isomerases. There are seven peptidyl-prolyl cis-trans isomerases in the rER, and so far, two of these enzymes, cyclophilin B and FKBP65, have been shown to be involved in collagen biosynthesis. The absence of either cyclophilin B or FKBP65 leads to a recessive form of osteogenesis imperfecta. The absence of FKBP22 leads to a kyphoscoliotic type of Ehlers-Danlos syndrome (EDS), and this type of EDS is classified as EDS type VI, which can also be caused by a deficiency in lysyl-hydroxylase 1. However, the lack of FKBP22 shows a wider spectrum of clinical phenotypes than the absence of lysyl-hydroxylase 1 and additionally includes myopathy, hearing loss, and aortic rupture. Here we show that FKBP22 catalyzes the folding of type III collagen and interacts with type III collagen, type VI collagen, and type X collagen, but not with type I collagen, type II collagen, or type V collagen. These restrictive interactions might help explain the broader phenotype observed in patients that lack FKBP22.


Subject(s)
Endoplasmic Reticulum, Rough/enzymology , Fibrillar Collagens/biosynthesis , Tacrolimus Binding Proteins/chemistry , Tacrolimus Binding Proteins/metabolism , Cyclophilins/genetics , Cyclophilins/metabolism , Endoplasmic Reticulum, Rough/metabolism , Fibrillar Collagens/chemistry , Humans , Protein Folding , Substrate Specificity , Tacrolimus/metabolism , Tacrolimus Binding Proteins/genetics
15.
J Biol Chem ; 288(44): 31437-46, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24043621

ABSTRACT

Collagen biosynthesis occurs in the rough endoplasmic reticulum, and many molecular chaperones and folding enzymes are involved in this process. The folding mechanism of type I procollagen has been well characterized, and protein disulfide isomerase (PDI) has been suggested as a key player in the formation of the correct disulfide bonds in the noncollagenous carboxyl-terminal and amino-terminal propeptides. Prolyl 3-hydroxylase 1 (P3H1) forms a hetero-trimeric complex with cartilage-associated protein and cyclophilin B (CypB). This complex is a multifunctional complex acting as a prolyl 3-hydroxylase, a peptidyl prolyl cis-trans isomerase, and a molecular chaperone. Two major domains are predicted from the primary sequence of P3H1: an amino-terminal domain and a carboxyl-terminal domain corresponding to the 2-oxoglutarate- and iron-dependent dioxygenase domains similar to the α-subunit of prolyl 4-hydroxylase and lysyl hydroxylases. The amino-terminal domain contains four CXXXC sequence repeats. The primary sequence of cartilage-associated protein is homologous to the amino-terminal domain of P3H1 and also contains four CXXXC sequence repeats. However, the function of the CXXXC sequence repeats is not known. Several publications have reported that short peptides containing a CXC or a CXXC sequence show oxido-reductase activity similar to PDI in vitro. We hypothesize that CXXXC motifs have oxido-reductase activity similar to the CXXC motif in PDI. We have tested the enzyme activities on model substrates in vitro using a GCRALCG peptide and the P3H1 complex. Our results suggest that this complex could function as a disulfide isomerase in the rough endoplasmic reticulum.


Subject(s)
Cyclophilins/chemistry , Endoplasmic Reticulum, Rough/chemistry , Extracellular Matrix Proteins/chemistry , Membrane Glycoproteins/chemistry , Multiprotein Complexes/chemistry , Protein Disulfide-Isomerases/chemistry , Proteins/chemistry , Proteoglycans/chemistry , Amino Acid Motifs , Animals , Chickens , Cyclophilins/genetics , Cyclophilins/metabolism , Endoplasmic Reticulum, Rough/genetics , Endoplasmic Reticulum, Rough/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Molecular Chaperones , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Prolyl Hydroxylases , Protein Disulfide-Isomerases/genetics , Protein Disulfide-Isomerases/metabolism , Protein Structure, Quaternary , Protein Structure, Tertiary , Proteins/genetics , Proteins/metabolism , Proteoglycans/genetics , Proteoglycans/metabolism
16.
J Biol Chem ; 288(26): 19166-76, 2013 Jun 28.
Article in English | MEDLINE | ID: mdl-23645670

ABSTRACT

Vascular Ehlers-Danlos syndrome (EDS) type IV is the most severe form of EDS. In many cases the disease is caused by a point mutation of Gly in type III collagen. A slower folding of the collagen helix is a potential cause for over-modifications. However, little is known about the rate of folding of type III collagen in patients with EDS. To understand the molecular mechanism of the effect of mutations, a system was developed for bacterial production of homotrimeric model polypeptides. The C-terminal quarter, 252 residues, of the natural human type III collagen was attached to (GPP)7 with the type XIX collagen trimerization domain (NC2). The natural collagen domain forms a triple helical structure without 4-hydroxylation of proline at a low temperature. At 33 °C, the natural collagenous part is denatured, but the C-terminal (GPP)7-NC2 remains intact. Switching to a low temperature triggers the folding of the type III collagen domain in a zipper-like fashion that resembles the natural process. We used this system for the two known EDS mutations (Gly-to-Val) in the middle at Gly-910 and at the C terminus at Gly-1018. In addition, wild-type and Gly-to-Ala mutants were made. The mutations significantly slow down the overall rate of triple helix formation. The effect of the Gly-to-Val mutation is much more severe compared with Gly-to-Ala. This is the first report on the folding of collagen with EDS mutations, which demonstrates local delays in the triple helix propagation around the mutated residue.


Subject(s)
Collagen Type III/genetics , Ehlers-Danlos Syndrome/genetics , Protein Folding , Circular Dichroism , Collagen/chemistry , Collagen Type III/chemistry , Humans , Mutation , Peptidylprolyl Isomerase/chemistry , Point Mutation , Protein Processing, Post-Translational , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Temperature , Trypsin/chemistry
17.
J Biol Chem ; 288(49): 35526-33, 2013 Dec 06.
Article in English | MEDLINE | ID: mdl-24169694

ABSTRACT

Decorin, the prototypical small leucine-rich proteoglycan, binds to collagen and thereby regulates collagen assembly into fibrils. The crystal structure of the decorin core protein revealed a tight dimer formed by the association of two monomers via their concave faces (Scott, P. G., McEwan, P. A., Dodd, C. M., Bergmann, E. M., Bishop, P. N., and Bella, J. (2004) Proc. Natl. Acad. Sci. U.S.A. 101, 15633-15638). Whether decorin binds collagen as a dimer has been controversial. Using analytical ultracentrifugation, we determined a dissociation constant of 1.37 ± 0.30 µm for the mouse decorin dimer. Dimerization could be abolished by engineering glycosylation sites into the dimer interface; other interface mutants remained dimeric. The monomeric mutants were as stable as wild-type decorin in thermal unfolding experiments. Mutations on the concave face of decorin abolished collagen binding regardless of whether the mutant proteins retained the ability to dimerize or not. We conclude that the concave face of decorin mediates collagen binding and that the dimer therefore must dissociate to bind collagen.


Subject(s)
Collagen/metabolism , Decorin/chemistry , Decorin/metabolism , Animals , Crystallography, X-Ray , Decorin/genetics , Glycosylation , HEK293 Cells , Humans , Mice , Models, Molecular , Mutagenesis, Site-Directed , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Stability , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
18.
J Biol Chem ; 288(34): 24742-52, 2013 Aug 23.
Article in English | MEDLINE | ID: mdl-23861401

ABSTRACT

Type I collagen extracted from tendon, skin, and bone of wild type and prolyl 3-hydroxylase 1 (P3H1) null mice shows distinct patterns of 3-hydroxylation and glycosylation of hydroxylysine residues. The A1 site (Pro-986) in the α1-chain of type I collagen is almost completely 3-hydroxylated in every tissue of the wild type mice. In contrast, no 3-hydroxylation of this proline residue was found in P3H1 null mice. Partial 3-hydroxylation of the A3 site (Pro-707) was present in tendon and bone, but absent in skin in both α-chains of the wild type animals. Type I collagen extracted from bone of P3H1 null mice shows a large reduction in 3-hydroxylation of the A3 site in both α-chains, whereas type I collagen extracted from tendon of P3H1 null mice shows little difference as compared with wild type. These results demonstrate that the A1 site in type I collagen is exclusively 3-hydroxylated by P3H1, and presumably, this enzyme is required for the 3-hydroxylation of the A3 site of both α-chains in bone but not in tendon. The increase in glycosylation of hydroxylysine in P3H1 null mice in bone was found to be due to an increased occupancy of normally glycosylated sites. Despite the severe disorganization of collagen fibrils in adult tissues, the D-period of the fibrils is unchanged. Tendon fibrils of newborn P3H1 null mice are well organized with only a slight increase in diameter. The absence of 3-hydroxyproline and/or the increased glycosylation of hydroxylysine in type I collagen disturbs the lateral growth of the fibrils.


Subject(s)
Collagen Type I/metabolism , Procollagen-Proline Dioxygenase/metabolism , Protein Processing, Post-Translational/physiology , Animals , Collagen Type I/genetics , Hydroxylation/physiology , Mice , Mice, Mutant Strains , Organ Specificity/physiology , Procollagen-Proline Dioxygenase/genetics , Proline/genetics , Proline/metabolism
19.
Biochim Biophys Acta ; 1833(11): 2479-91, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23602968

ABSTRACT

Extracellular matrix (ECM) proteins create structural frameworks in tissues such as bone, skin, tendon and cartilage etc. These connective tissues play important roles in the development and homeostasis of organs. Collagen is the most abundant ECM protein and represents one third of all proteins in humans. The biosynthesis of ECM proteins occurs in the rough endoplasmic reticulum (rER). This review describes the current understanding of the biosynthesis and folding of procollagens, which are the precursor molecules of collagens, in the rER. Multiple folding enzymes and molecular chaperones are required for procollagen to establish specific posttranslational modifications, and facilitate folding and transport to the cell surface. Thus, this molecular ensemble in the rER contributes to ECM maturation and to the development and homeostasis of tissues. Mutations in this ensemble are likely candidates for connective tissue disorders. This article is part of a Special Issue entitled: Functional and structural diversity of endoplasmic reticulum.


Subject(s)
Endoplasmic Reticulum, Rough/metabolism , Extracellular Matrix Proteins/metabolism , Molecular Chaperones/metabolism , Procollagen/metabolism , Animals , Humans
20.
J Biol Chem ; 287(53): 44536-45, 2012 Dec 28.
Article in English | MEDLINE | ID: mdl-23132862

ABSTRACT

Precise mapping and unraveling the mechanism of interaction or degradation of a certain type of collagen triple helix requires the generation of short and stable collagenous fragments. This is a great challenge especially for hetero-trimeric collagens, where chain composition and register (stagger) are important factors. No system has been reported that can be efficiently used to generate a natural collagenous fragment with exact chain composition and desired chain register. The NC2 domain (only 35-50 residues) of FACIT collagens is a potent trimerization domain. In the case of type IX collagen it provides the efficient selection and hetero-trimerization of three distinct chains. The ability of the NC2 domain to determine the chain register of the triple helix is studied. We generated three possible sequence combinations (α1α1α2, α1α2α1, α2α1α1) of a type I collagen fragment (the binding region for the von Willebrand factor A3 domain) attached to the NC2 domain. In addition, two control combinations were produced that constitute homo-trimers of (α1)(3) or (α2)(3). For the hetero-trimeric constructs, α1α1α2 demonstrated a higher melting temperature than the other two. Binding experiments with the von Willebrand factor A3 domain revealed the homo-trimer of (α1)(3) as the strongest binding construct, whereas the homo-trimer of (α2)(3) showed no binding. For hetero-trimers, α1α1α2 was found to be the strongest binding construct. Differences in thermal stability and binding to the A3 domain unambiguously demonstrate that the NC2 domain of type IX collagen determines not only the chain composition but also the chain register of the adjacent triple helix.


Subject(s)
Collagen Type IX/chemistry , Protein Multimerization , Amino Acid Sequence , Collagen Type IX/genetics , Collagen Type IX/metabolism , Crystallography, X-Ray , Humans , Molecular Sequence Data , Protein Binding , Protein Folding , Protein Stability , Protein Structure, Secondary , Protein Structure, Tertiary , von Willebrand Factor/genetics , von Willebrand Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL