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










Publication year range
1.
Sci Rep ; 13(1): 17039, 2023 10 09.
Article in English | MEDLINE | ID: mdl-37814009

ABSTRACT

Amniotic fluid is a complex biological medium that offers protection to the fetus and plays a key role in normal fetal nutrition, organogenesis, and potentially fetal programming. Amniotic fluid is also critically involved in longitudinally shaping the in utero milieu during pregnancy. Yet, the molecular mechanism(s) of action by which amniotic fluid regulates fetal development is ill-defined partly due to an incomplete understanding of the evolving composition of the amniotic fluid proteome. Prior research consisting of cross-sectional studies suggests that the amniotic fluid proteome changes as pregnancy advances, yet longitudinal alterations have not been confirmed because repeated sampling is prohibitive in humans. We therefore performed serial amniocenteses at early, mid, and late gestational time-points within the same pregnancies in a rhesus macaque model. Longitudinally-collected rhesus amniotic fluid samples were paired with gestational-age matched cross-sectional human samples. Utilizing LC-MS/MS isobaric labeling quantitative proteomics, we demonstrate considerable cross-species similarity between the amniotic fluid proteomes and large scale gestational-age associated changes in protein content throughout pregnancy. This is the first study to compare human and rhesus amniotic fluid proteomic profiles across gestation and establishes a reference amniotic fluid proteome. The non-human primate model holds promise as a translational platform for amniotic fluid studies.


Subject(s)
Amniotic Fluid , Proteome , Female , Animals , Humans , Pregnancy , Amniotic Fluid/metabolism , Macaca mulatta/metabolism , Proteome/metabolism , Chromatography, Liquid , Proteomics , Cross-Sectional Studies , Tandem Mass Spectrometry , Gestational Age
3.
Matrix Biol ; 123: 17-33, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37683955

ABSTRACT

Although abnormal TGFß signaling is observed in several heritable forms of thoracic aortic aneurysms and dissections including Marfan syndrome, its precise role in aortic disease progression is still disputed. Using a mouse genetic approach and quantitative isobaric labeling proteomics, we sought to elucidate the role of TGFß signaling in three Fbn1 mutant mouse models representing a range of aortic disease from microdissection (without aneurysm) to aneurysm (without rupture) to aneurysm and rupture. Results indicated that reduced TGFß signaling and increased mast cell proteases were associated with microdissection. In contrast, increased abundance of extracellular matrix proteins, which could be reporters for positive TGFß signaling, were associated with aneurysm. Marked reductions in collagens and fibrillins, and increased TGFß signaling, were associated with aortic rupture. Our data indicate that TGFß signaling performs context-dependent roles in the pathogenesis of thoracic aortic disease.


Subject(s)
Aortic Aneurysm, Thoracic , Marfan Syndrome , Humans , Aortic Aneurysm, Thoracic/genetics , Fibrillin-1/genetics , Fibrillins , Marfan Syndrome/genetics , Marfan Syndrome/pathology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
4.
Exp Eye Res ; 234: 109605, 2023 09.
Article in English | MEDLINE | ID: mdl-37506755

ABSTRACT

PURPOSE: Biophysical and biochemical attributes of the extracellular matrix are major determinants of cell fate in homeostasis and disease. Ocular hypertension and glaucoma are diseases where the trabecular meshwork tissue responsible for aqueous humor egress becomes stiffer accompanied by changes in its matrisome in a segmental manner with regions of high or low flow. Prior studies demonstrate these alterations in the matrix are dynamic in response to age and pressure changes. The underlying reason for segmentation or differential response to pressure and stiffening are unknown. This is largely due to a lack of appropriate models (in vitro or ex vivo) to study this phenomena. METHODS: Primary trabecular meshwork cells were isolated from segmental flow regions, and cells were cultured for 4 weeks in the presence or absence or dexamethasone to obtain cell derived matrices (CDM). The biomechanical attributes of the CDM, composition of the matrisome, and incidence of crosslinks were determined by atomic force microscopy and mass spectrometry. RESULTS: Data demonstrate that matrix deposited by cells from low flow regions are stiffer and exhibit a greater number of immature and mature crosslinks, and that these are exacerbated in the presence of steroid. We also show a differential response of high or low flow cells to steroid via changes observed in the matrix composition. However, no correlations were observed between elastic moduli and presence or absence of mature and immature crosslinks in the CDMs. CONCLUSION: Regardless of a direct correlation between matrix stiffness and crosslinks, we observed distinct differences in the composition and mechanics of the matrices deposited by segmental flow cells. These results suggest distinct differences in cellular identify and likely a basis for mechanical memory post isolation and culture. Nevertheless, we conclude that although a mechanistic basis for matrix stiffness was undetermined in this study, it is a viable tool to study cell-matrix interactions and further our understanding of trabecular meshwork pathobiology.


Subject(s)
Glaucoma , Ocular Hypertension , Humans , Trabecular Meshwork , Extracellular Matrix , Aqueous Humor
5.
bioRxiv ; 2023 Mar 12.
Article in English | MEDLINE | ID: mdl-36945588

ABSTRACT

Biophysical and biochemical attributes of the extracellular matrix are major determinants of cell fate in homeostasis and disease. Ocular hypertension and glaucoma are diseases where the trabecular meshwork tissue responsible for aqueous humor egress becomes stiffer accompanied by changes in its matrisome in a segmental manner with regions of high or low flow. Prior studies demonstrate these alterations in the matrix are dynamic in response to age and pressure changes. The underlying reason for segmentation or differential response to pressure and stiffening are unknown. This is largely due to a lack of appropriate models ( in vitro or ex vivo ) to study this phenomena. In this study, we characterize the biomechanical attributes, matrisome, and incidence of crosslinks in the matrix deposited by primary cells isolated from segmental flow regions and when treated with glucocorticosteroid. Data demonstrate that matrix deposited by cells from low flow regions are stiffer and exhibit a greater number of immature and mature crosslinks, and that these are exacerbated in the presence of steroid. We also show a differential response of high or low flow cells to steroid via changes observed in the matrix composition. We conclude that although a mechanistic basis for matrix stiffness was undetermined in this study, it is a viable tool to study cell-matrix interactions and further our understanding of trabecular meshwork pathobiology.

6.
Exp Eye Res ; 227: 109358, 2023 02.
Article in English | MEDLINE | ID: mdl-36572168

ABSTRACT

The α-crystallin small heat shock proteins contribute to the transparency and refractive properties of the vertebrate eye lens and prevent the protein aggregation that would otherwise produce lens cataracts, the leading cause of human blindness. There are conflicting data in the literature as to what role the α-crystallins may play in early lens development. In this study, we used CRISPR gene editing to produce zebrafish lines with mutations in each of the three α-crystallin genes (cryaa, cryaba and cryabb) to prevent protein production. The absence of each α-crystallin protein was analyzed by mass spectrometry, and lens phenotypes were assessed with differential interference contrast microscopy and histology. Loss of αA-crystallin produced a variety of lens defects with varying severity in larvae at 3 and 4 dpf but little substantial change in normal fiber cell denucleation. Loss of αBa-crystallin produced no substantial lens defects. Our cryabb mutant produced a truncated αBb-crystallin protein and showed no substantial change in lens development. Mutation of each α-crystallin gene did not alter the mRNA levels of the remaining two, suggesting a lack of genetic compensation. These data suggest that αA-crystallin plays some role in lens development, but the range of phenotype severity in null mutants indicates its loss simply increases the chance for defects and that the protein is not essential. Our finding that cryaba and cryabb mutants lack noticeable lens defects is congruent with insubstantial transcript levels for these genes in lens epithelial and fiber cells through five days of development. Future experiments can explore the molecular mechanisms leading to lens defects in cryaa null mutants and the impact of αA-crystallin loss during zebrafish lens aging.


Subject(s)
Cataract , Crystallins , Lens, Crystalline , alpha-Crystallin A Chain , alpha-Crystallins , Animals , Humans , Zebrafish , alpha-Crystallins/genetics , alpha-Crystallins/metabolism , Crystallins/genetics , Crystallins/metabolism , alpha-Crystallin A Chain/metabolism , Lens, Crystalline/metabolism , Proteins/metabolism , Cataract/metabolism
7.
Blood Adv ; 7(8): 1366-1378, 2023 04 25.
Article in English | MEDLINE | ID: mdl-36219587

ABSTRACT

Low-density lipoprotein (LDL) contributes to atherogenesis and cardiovascular disease through interactions with peripheral blood cells, especially platelets. However, mechanisms by which LDL affects platelet activation and atherothrombosis, and how to best therapeutically target and safely prevent such responses remain unclear. Here, we investigate how oxidized low-density lipoprotein (oxLDL) enhances glycoprotein VI (GPVI)-mediated platelet hemostatic and procoagulant responses, and how traditional and emerging antiplatelet therapies affect oxLDL-enhanced platelet procoagulant activity ex vivo. Human platelets were treated with oxLDL and the GPVI-specific agonist, crosslinked collagen-related peptide, and assayed for hemostatic and procoagulant responses in the presence of inhibitors of purinergic receptors (P2YR), cyclooxygenase (COX), and tyrosine kinases. Ex vivo, oxLDL enhanced GPVI-mediated platelet dense granule secretion, α-granule secretion, integrin activation, thromboxane generation and aggregation, as well as procoagulant phosphatidylserine exposure and fibrin generation. Studies of washed human platelets, as well as platelets from mouse and nonhuman primate models of hyperlipidemia, further determined that P2YR antagonists (eg, ticagrelor) and Bruton tyrosine kinase inhibitors (eg, ibrutinib) reduced oxLDL-mediated platelet responses and procoagulant activity, whereas COX inhibitors (eg, aspirin) had no significant effect. Together, our results demonstrate that oxLDL enhances GPVI-mediated platelet procoagulant activity in a manner that may be more effectively reduced by P2YR antagonists and tyrosine kinase inhibitors compared with COX inhibitors.


Subject(s)
Hemostatics , Platelet Aggregation Inhibitors , Humans , Mice , Animals , Platelet Aggregation Inhibitors/pharmacology , Lipoproteins, LDL/pharmacology
8.
Vasc Endovascular Surg ; 56(3): 244-252, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34961389

ABSTRACT

OBJECTIVE: Tobacco smoke exposure is a major risk factor for aortic aneurysm development. However, the initial aortic response to tobacco smoke, preceding aneurysm formation, is not well understood. We sought to create a model to determine the effect of solubilized tobacco smoke (STS) on the thoracic and abdominal aorta of mice as well as on cultured human aortic smooth muscle cells (HASMCs). METHODS: Tobacco smoke was solubilized and delivered to mice via implanted osmotic minipumps. Twenty male C57BL/6 mice received STS or vehicle infusion. The descending thoracic, suprarenal abdominal, and infrarenal abdominal segments of the aorta were assessed for elastic lamellar damage, smooth muscle cell phenotype, and infiltration of inflammatory cells. Cultured HASMCs grown in media containing STS were compared to cells grown in standard media in order to verify our in vivo findings. RESULTS: Tobacco smoke solution caused significantly more breaks in the elastic lamellae of the thoracic and abdominal aorta compared to control solution (P< .0001) without inciting an inflammatory infiltrate. Elastin breaks occurred more frequently in the abdominal aorta than the thoracic aorta (P < .01). Exposure to STS-induced aortic microdissections and downregulation of α-smooth muscle actin (α-SMA) by vascular smooth muscle cells (VSMCs). Treatment of cultured HASMCs with STS confirmed the decrease in α-SMA expression. CONCLUSION: Delivery of STS via osmotic minipumps appears to be a promising model for investigating the early aortic response to tobacco smoke exposure. The initial effect of tobacco smoke exposure on the aorta is elastic lamellar damage and downregulation of (α-SMA) expression by VSMCs. Elastic lamellar damage occurs more frequently in the abdominal aorta than the thoracic aorta and does not seem to be mediated by the presence of macrophages or other inflammatory cells.


Subject(s)
Aortic Aneurysm, Abdominal , Tobacco Smoke Pollution , Animals , Aorta, Abdominal , Aortic Aneurysm, Abdominal/chemically induced , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle/metabolism , Nicotiana , Tobacco Smoke Pollution/adverse effects , Treatment Outcome
9.
Invest Ophthalmol Vis Sci ; 62(15): 27, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34964803

ABSTRACT

Purpose: Exfoliation syndrome (XFS) is a condition characterized by the production of insoluble fibrillar aggregates (exfoliation material; XFM) in the eye and elsewhere. Many patients with XFS progress to exfoliation glaucoma (XFG), a significant cause of global blindness. We used quantitative mass spectrometry to analyze the composition of XFM in lens capsule specimens and in aqueous humor (AH) samples from patients with XFS, patients with XFG and unaffected individuals. Methods: Pieces of lens capsule and samples of AH were obtained with consent from patients undergoing cataract surgery. Tryptic digests of capsule or AH were analyzed by high-performance liquid chromatography-mass spectrometry and relative differences between samples were quantified using the tandem mass tag technique. The distribution of XFM on the capsular surface was visualized by SEM and super-resolution light microscopy. Results: A small set of proteins was consistently upregulated in capsule samples from patients with XFS and patients with XFG, including microfibril components fibrillin-1, latent transforming growth factor-ß-binding protein-2 and latent transforming growth factor-ß-binding protein-3. Lysyl oxidase-like 1, a cross-linking enzyme associated with XFS in genetic studies, was an abundant XFM constituent. Ligands of the transforming growth factor-ß superfamily were prominent, including LEFTY2, a protein best known for its role in establishing the embryonic body axis. Elevated levels of LEFTY2 were also detected in AH from patients with XFG, a finding confirmed subsequently by ELISA. Conclusions: This analysis verified the presence of suspected XFM proteins and identified novel components. Quantitative comparisons between patient samples revealed a consistent XFM proteome characterized by strong expression of fibrillin-1, lysyl oxidase-like-1, and LEFTY2. Elevated levels of LEFTY2 in the AH of patients with XFG may serve as a biomarker for the disease.


Subject(s)
Aqueous Humor/metabolism , Crystallins/metabolism , Exfoliation Syndrome/metabolism , Glaucoma, Open-Angle/metabolism , Lens Capsule, Crystalline/metabolism , Protein Aggregates/physiology , Aged , Aged, 80 and over , Amino Acid Oxidoreductases/metabolism , Chromatography, High Pressure Liquid , Crystallins/ultrastructure , Enzyme-Linked Immunosorbent Assay , Female , Fibrillin-1/metabolism , Fluorescent Antibody Technique, Indirect , Humans , Latent TGF-beta Binding Proteins/metabolism , Left-Right Determination Factors/metabolism , Lens Capsule, Crystalline/ultrastructure , Male , Mass Spectrometry , Microscopy, Electron, Scanning , Middle Aged
10.
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
11.
Sci Signal ; 11(557)2018 11 20.
Article in English | MEDLINE | ID: mdl-30459282

ABSTRACT

Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma of childhood with an unmet clinical need for decades. A single oncogenic fusion gene is associated with treatment resistance and a 40 to 45% decrease in overall survival. We previously showed that expression of this PAX3:FOXO1 fusion oncogene in alveolar RMS (aRMS) mediates tolerance to chemotherapy and radiotherapy and that the class I-specific histone deacetylase (HDAC) inhibitor entinostat reduces PAX3:FOXO1 protein abundance. Here, we established the antitumor efficacy of entinostat with chemotherapy in various preclinical cell and mouse models and found that HDAC3 inhibition was the primary mechanism of entinostat-induced suppression of PAX3:FOXO1 abundance. HDAC3 inhibition by entinostat decreased the activity of the chromatin remodeling enzyme SMARCA4, which, in turn, derepressed the microRNA miR-27a. This reexpression of miR-27a led to PAX3:FOXO1 mRNA destabilization and chemotherapy sensitization in aRMS cells in culture and in vivo. Furthermore, a phase 1 clinical trial (ADVL1513) has shown that entinostat is tolerable in children with relapsed or refractory solid tumors and is planned for phase 1B cohort expansion or phase 2 clinical trials. Together, these results implicate an HDAC3-SMARCA4-miR-27a-PAX3:FOXO1 circuit as a driver of chemoresistant aRMS and suggest that targeting this pathway with entinostat may be therapeutically effective in patients.


Subject(s)
DNA Helicases/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , MicroRNAs/metabolism , Nuclear Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Paired Box Transcription Factors/metabolism , Rhabdomyosarcoma, Alveolar/metabolism , Transcription Factors/metabolism , Animals , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Cell Line, Tumor , Computational Biology , Drug Resistance, Neoplasm , Epigenesis, Genetic , Female , Fluorescence Resonance Energy Transfer , Forkhead Box Protein O1/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Mice , Neoplasm Transplantation , PAX3 Transcription Factor/metabolism , Pyridines/pharmacology , Sequence Analysis, RNA , Vincristine/pharmacology
12.
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
13.
Blood ; 125(9): 1488-96, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-25587039

ABSTRACT

Activation of coagulation factor XI (FXI) may play a role in hemostasis. The primary substrate of activated FXI (FXIa) is FIX, leading to FX activation (FXa) and thrombin generation. However, recent studies suggest the hemostatic role of FXI may not be restricted to the activation of FIX. We explored whether FXI could interact with and inhibit the activity of tissue factor pathway inhibitor (TFPI). TFPI is an essential reversible inhibitor of activated factor X (FXa) and also inhibits the FVIIa-TF complex. We found that FXIa neutralized both endothelium- and platelet-derived TFPI by cleaving the protein between the Kunitz (K) 1 and K2 domains (Lys86/Thr87) and at the active sites of the K2 (Arg107/Gly108) and K3 (Arg199/Ala200) domains. Addition of FXIa to plasma was able to reverse the ability of TFPI to prolong TF-initiated clotting times in FXI- or FIX-deficient plasma, as well as FXa-initiated clotting times in FX-deficient plasma. Treatment of cultured endothelial cells with FXIa increased the generation of FXa and promoted TF-dependent fibrin formation in recalcified plasma. Together, these results suggest that the hemostatic role of FXIa may be attributed not only to activation of FIX but also to promoting the extrinsic pathway of thrombin generation through inactivation of TFPI.


Subject(s)
Blood Coagulation/physiology , Blood Platelets/metabolism , Factor IX/metabolism , Factor XIa/metabolism , Factor Xa/metabolism , Fibrin/metabolism , Lipoproteins/metabolism , Blood Platelets/cytology , Blotting, Western , Cells, Cultured , Flow Cytometry , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Lipoproteins/genetics , Mutation/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
14.
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
15.
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
16.
J Biol Chem ; 287(26): 22253-65, 2012 Jun 22.
Article in English | MEDLINE | ID: mdl-22556420

ABSTRACT

The rate-limiting step of folding of the collagen triple helix is catalyzed by cyclophilin B (CypB). The G6R mutation in cyclophilin B found in the American Quarter Horse leads to autosomal recessive hyperelastosis cutis, also known as hereditary equine regional dermal asthenia. The mutant protein shows small structural changes in the region of the mutation at the side opposite the catalytic domain of CypB. The peptidylprolyl cis-trans isomerase activity of the mutant CypB is normal when analyzed in vitro. However, the biosynthesis of type I collagen in affected horse fibroblasts shows a delay in folding and secretion and a decrease in hydroxylysine and glucosyl-galactosyl hydroxylysine. This leads to changes in the structure of collagen fibrils in tendon, similar to those observed in P3H1 null mice. In contrast to cyclophilin B null mice, where little 3-hydroxylation was found in type I collagen, 3-hydroxylation of type I collagen in affected horses is normal. The mutation disrupts the interaction of cyclophilin B with the P-domain of calreticulin, with lysyl hydroxylase 1, and probably other proteins, such as the formation of the P3H1·CypB·cartilage-associated protein complex, resulting in less effective catalysis of the rate-limiting step in collagen folding in the rough endoplasmic reticulum.


Subject(s)
Collagen/chemistry , Cyclophilins/genetics , Mutation , Peptidylprolyl Isomerase/chemistry , Skin Diseases/genetics , Skin Diseases/veterinary , cis-trans-Isomerases/metabolism , Animals , Asthenia , Circular Dichroism , Endoplasmic Reticulum, Rough/metabolism , Horses , Kinetics , Mice , Mice, Transgenic , Molecular Chaperones/metabolism , Protein Binding , Protein Folding , Protein Structure, Tertiary , Surface Plasmon Resonance
17.
J Biol Chem ; 285(31): 23721-31, 2010 Jul 30.
Article in English | MEDLINE | ID: mdl-20507993

ABSTRACT

The mechanism of chain selection and trimerization of fibril-associated collagens with interrupted triple helices (FACITs) differs from that of fibrillar collagens that have special C-propeptides. We recently showed that the second carboxyl-terminal non-collagenous domain (NC2) of homotrimeric collagen XIX forms a stable trimer and substantially stabilizes a collagen triple helix attached to either end. We then hypothesized a general trimerizing role for the NC2 domain in other FACITs. Here we analyzed the NC2 domain of human heterotrimeric collagen IX, the only member of FACITs with all three chains encoded by distinct genes. Upon oxidative folding of equimolar amounts of the alpha1, alpha2, and alpha3 chains of NC2, a stable heterotrimer with a disulfide bridge between alpha1 and alpha3 chains is formed. Our experiments show that this heterotrimerization domain can stabilize a short triple helix attached at the carboxyl-terminal end and allows for the proper oxidation of the cystine knot of type III collagen after the short triple helix.


Subject(s)
Collagen Type IX/chemistry , Amino Acid Sequence , C-Peptide/chemistry , Circular Dichroism , Cloning, Molecular , Dimerization , Humans , Molecular Sequence Data , Oxygen/chemistry , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Sequence Homology, Amino Acid , Thermodynamics , Thrombin/chemistry
18.
J Biol Chem ; 285(22): 17253-62, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20363744

ABSTRACT

Osteogenesis imperfecta (OI) is a skeletal disorder primarily caused by mutations in the type I collagen genes. However, recent investigations have revealed that mutations in the genes encoding for cartilage-associated protein (CRTAP) or prolyl 3-hydroxylase 1 (P3H1) can cause a severe, recessive form of OI. These reports show minimal 3-hydroxylation of key proline residues in type I collagen as a result of CRTAP or P3H1 deficiency and demonstrate the importance of P3H1 and CRTAP to bone structure and development. P3H1 and CRTAP have previously been shown to form a stable complex with cyclophilin B, and P3H1 was shown to catalyze the 3-hydroxylation of specific proline residues in procollagen I in vitro. Here we describe a mouse model in which the P3H1 gene has been inactivated. Our data demonstrate abnormalities in collagen fibril ultrastructure in tendons from P3H1 null mice by electron microscopy. Differences are also seen in skin architecture, as well as in developing limbs by histology. Additionally bone mass and strength were significantly lower in the P3H1 mice as compared with wild-type littermates. Altogether these investigations demonstrate disturbances of collagen fiber architecture in tissues rich in fibrillar collagen, including bone, tendon, and skin. This model system presents a good opportunity to study the underlying mechanisms of recessive OI and to better understand its effects in humans.


Subject(s)
Bone and Bones/metabolism , Collagen/chemistry , Procollagen-Proline Dioxygenase/genetics , Procollagen-Proline Dioxygenase/physiology , Skin/metabolism , Tendons/metabolism , Animals , Bone and Bones/embryology , Bone and Bones/ultrastructure , Extracellular Matrix/metabolism , Gene Expression Regulation, Developmental , Hydrolysis , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Electron/methods , Osteogenesis Imperfecta/metabolism , Protein Processing, Post-Translational , Skin/embryology , Skin/ultrastructure , Tendons/embryology , Tendons/ultrastructure
19.
J Vet Intern Med ; 21(5): 1067-75, 2007.
Article in English | MEDLINE | ID: mdl-17939566

ABSTRACT

BACKGROUND: Transdermal fentanyl is used clinically in horses based on pharmacokinetic data and antinociceptive effects documented in other species. HYPOTHESIS: Fentanyl IV administration increases both visceral and somatic nociceptive threshold in conscious horses. ANIMALS: Six clinically normal horses, each fitted with a permanent gastric cannula. METHODS: Visceral nociception was evaluated with 2 methods of threshold detection--olorectal distention and duodenal distention. Somatic nociception was assessed by measurement of thermal threshold. Fentanyl was administered as an increasing stepwise infusion followed by a continuous-rate infusion for a total of 2 hours. There were 4 doses of fentanyl and 1 dose each of saline and xylazine administered to each horse. Serum fentanyl concentrations were measured and the resulting data were used to determine pharmacokinetic parameters for each horse. All data were analyzed by means of a 3-factor analysis of variance followed by either a simple t test or a Bonferroni t test for multiple comparisons. RESULTS: Fentanyl administration did not result in significant changes in duodenal or colorectal distention threshold. Thermal threshold showed an increased trend at the 15-minute time point for the highest fentanyl group only, with a corresponding mean serum fentanyl concentration of 7.82 +/- 2.10 ng/mL. Two horses in this group became agitated and tachycardic during the first 15 minutes of the infusion. CONCLUSIONS AND CLINICAL IMPORTANCE: Fentanyl did not produce a significant antinociceptive effect at the doses used, 2 of which resulted in serum concentrations above the nociceptive threshold in other species.


Subject(s)
Analgesics, Opioid/pharmacology , Fentanyl/pharmacology , Horses/physiology , Pain Measurement/drug effects , Pain Measurement/veterinary , Analgesics, Opioid/pharmacokinetics , Analysis of Variance , Animals , Body Temperature/drug effects , Colon/drug effects , Female , Fentanyl/pharmacokinetics , Gastrointestinal Motility/drug effects , Heart Rate/drug effects , Horses/metabolism , Injections, Intravenous/veterinary , Male , Random Allocation , Respiration/drug effects , Stomach/drug effects
20.
J Anal Toxicol ; 31(2): 87-92, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17536743

ABSTRACT

A method for the extraction and quantitation of procaine in equine plasma was developed for use with liquid chromatography-mass spectrometry (LC-MS). Procaine was isolated from equine plasma by liquid-liquid extraction at pH 11 with dichloromethane using procaine-d10 as an internal standard. Quantitation was achieved by LC-MS using a 3-microm C-18 column coupled to an electrospray ionization source on a linear ion-trap mass spectrometer. The limit of detection and limit of quantitation was determined to be 50 and 200 pg/mL, respectively. The lowest limit of detection determined by previous methods was 1 ng/mL. Administration samples were obtained as part of a larger study to determine a regulatory limit for procaine in racehorses and procaine concentrations were determined using this method.


Subject(s)
Anesthetics, Local/blood , Chromatography, High Pressure Liquid , Procaine/blood , Spectrometry, Mass, Electrospray Ionization/methods , Substance Abuse Detection/methods , Anesthetics, Local/pharmacokinetics , Animals , Doping in Sports , Female , Horses , Injections, Subcutaneous , Male , Nerve Block/methods , Procaine/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...