Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 176
Filter
1.
FASEB J ; 35(3): e21399, 2021 03.
Article in English | MEDLINE | ID: mdl-33559227

ABSTRACT

The high-mobility group box-1 (HMGB1) protein is a transcription-regulating protein located in the nucleus. However, it serves as a damage-associated molecular pattern protein that activates immune cells and stimulates inflammatory cytokines to accentuate neuroinflammation after release from damaged cells. In contrast, Inter-alpha Inhibitor Proteins (IAIPs) are proteins with immunomodulatory effects including inhibition of pro-inflammatory cytokines. We have demonstrated that IAIPs exhibit neuroprotective properties in neonatal rats exposed to hypoxic-ischemic (HI) brain injury. In addition, previous studies have suggested that the light chain of IAIPs, bikunin, may exert its anti-inflammatory effects by inhibiting HMGB1 in a variety of different injury models in adult subjects. The objectives of the current study were to confirm whether HMGB1 is a target of IAIPs by investigating the potential binding characteristics of HMGB1 and IAIPs in vitro, and co-localization in vivo in cerebral cortices after exposure to HI injury. Solid-phase binding assays and surface plasmon resonance (SPR) were used to determine the physical binding characteristics between IAIPs and HMGB1. Cellular localizations of IAIPs-HMGB1 in neonatal rat cortex were visualized by double labeling with anti-IAIPs and anti-HMGB1 antibodies. Solid-phase binding and SPR demonstrated specific binding between IAIPs and HMGB1 in vitro. Cortical cytoplasmic and nuclear co-localization of IAIPs and HMGB1 were detected by immunofluorescent staining in control and rats immediately and 3 hours after HI. In conclusion, HMGB1 and IAIPs exhibit direct binding in vitro and co-localization in vivo in neonatal rats exposed to HI brain injury suggesting HMGB1 could be a target of IAIPs.


Subject(s)
Alpha-Globulins/chemistry , Cerebral Cortex/chemistry , HMGB1 Protein/chemistry , Hypoxia-Ischemia, Brain/metabolism , Alpha-Globulins/analysis , Animals , Animals, Newborn , Female , Fluorescent Antibody Technique , HMGB1 Protein/analysis , Immunohistochemistry , Rats , Rats, Wistar , Surface Plasmon Resonance
2.
Int J Mol Sci ; 21(23)2020 Dec 02.
Article in English | MEDLINE | ID: mdl-33276548

ABSTRACT

Perinatal hypoxia-ischemia (HI) is a major cause of brain injury and mortality in neonates. Hypoxic-ischemic encephalopathy (HIE) predisposes infants to long-term cognitive deficits that influence their quality of life and place a large burden on society. The only approved treatment to protect the brain after HI is therapeutic hypothermia, which has limited effectiveness, a narrow therapeutic time window, and is not considered safe for treatment of premature infants. Alternative or adjunctive therapies are needed to improve outcomes of full-term and premature infants after exposure to HI. Inter-alpha inhibitor proteins (IAIPs) are immunomodulatory molecules that are proposed to limit the progression of neonatal inflammatory conditions, such as sepsis. Inflammation exacerbates neonatal HIE and suggests that IAIPs could attenuate HI-related brain injury and improve cognitive outcomes associated with HIE. Recent studies have shown that intraperitoneal treatment with IAIPs can decrease neuronal and non-neuronal cell death, attenuate glial responses and leukocyte invasion, and provide long-term behavioral benefits in neonatal rat models of HI-related brain injury. The present review summarizes these findings and outlines the remaining experimental analyses necessary to determine the clinical applicability of this promising neuroprotective treatment for neonatal HI-related brain injury.


Subject(s)
Brain Injuries/drug therapy , Hypoxia-Ischemia, Brain/drug therapy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Alpha-Globulins/chemistry , Alpha-Globulins/genetics , Alpha-Globulins/metabolism , Animals , Brain Injuries/diagnosis , Brain Injuries/etiology , Brain Injuries/metabolism , Disease Management , Disease Susceptibility , Humans , Hypoxia-Ischemia, Brain/diagnosis , Hypoxia-Ischemia, Brain/etiology , Hypoxia-Ischemia, Brain/metabolism , Infant, Newborn , Neurons/metabolism , Neuroprotection , Structure-Activity Relationship
3.
J Biol Chem ; 295(16): 5278-5291, 2020 04 17.
Article in English | MEDLINE | ID: mdl-32144206

ABSTRACT

Inter-α-inhibitor is a proteoglycan essential for mammalian reproduction and also plays a less well-characterized role in inflammation. It comprises two homologous "heavy chains" (HC1 and HC2) covalently attached to chondroitin sulfate on the bikunin core protein. Before ovulation, HCs are transferred onto the polysaccharide hyaluronan (HA) to form covalent HC·HA complexes, thereby stabilizing an extracellular matrix around the oocyte required for fertilization. Additionally, such complexes form during inflammatory processes and mediate leukocyte adhesion in the synovial fluids of arthritis patients and protect against sepsis. Here using X-ray crystallography, we show that human HC1 has a structure similar to integrin ß-chains, with a von Willebrand factor A domain containing a functional metal ion-dependent adhesion site (MIDAS) and an associated hybrid domain. A comparison of the WT protein and a variant with an impaired MIDAS (but otherwise structurally identical) by small-angle X-ray scattering and analytical ultracentrifugation revealed that HC1 self-associates in a cation-dependent manner, providing a mechanism for HC·HA cross-linking and matrix stabilization. Surprisingly, unlike integrins, HC1 interacted with RGD-containing ligands, such as fibronectin, vitronectin, and the latency-associated peptides of transforming growth factor ß, in a MIDAS/cation-independent manner. However, HC1 utilizes its MIDAS motif to bind to and inhibit the cleavage of complement C3, and small-angle X-ray scattering-based modeling indicates that this occurs through the inhibition of the alternative pathway C3 convertase. These findings provide detailed structural and functional insights into HC1 as a regulator of innate immunity and further elucidate the role of HC·HA complexes in inflammation and ovulation.


Subject(s)
Alpha-Globulins/chemistry , Extracellular Matrix/metabolism , Immunity, Innate , Molecular Dynamics Simulation , Ovulation , Humans , Integrin beta Chains/chemistry , Protein Domains , von Willebrand Factor/chemistry
4.
J Biol Chem ; 294(42): 15495-15504, 2019 10 18.
Article in English | MEDLINE | ID: mdl-31484722

ABSTRACT

Destruction of the cartilage matrix in joints is an important feature of arthritis. Proteolytic degradation of cartilage glycoproteins can contribute to the loss of matrix integrity. Human inter-α-inhibitor (IαI), which stabilizes the extracellular matrix, is composed of the light-chain serine proteinase inhibitor bikunin and two homologous heavy chains (HC1 and HC2) covalently linked through chondroitin 4-sulfate. Inflammation promotes the transfer of HCs from chondroitin 4-sulfate to hyaluronan by tumor necrosis factor-stimulated gene-6 protein (TSG-6). This reaction generates a covalent complex between the heavy chains and hyaluronan that can promote leukocyte invasion. This study demonstrates that both IαI and the HC-hyaluronan complex are substrates for the extracellular matrix proteases ADAMTS-5 and matrix metalloprotease (MMP) -3, -7, and -13. The major cleavage sites for all four proteases are found in the C terminus of HC2. ADAMTS-5 and MMP-7 displayed the highest activity toward HC2. ADAMTS-5 degradation products were identified in mass spectrometric analysis of 29 of 33 arthropathic patients, indicating that ADAMTS-5 cleavage occurs in synovial fluid in arthritis. After cleavage, free HC2, together with TSG-6, is able to catalyze the transfer of heavy chains to hyaluronan. The release of extracellular matrix bound HC2 is likely to increase the mobility of the HC2/TSG-6 catalytic unit and consequently increase the rate of the HC transfer reaction. Ultimately, ADAMTS-5 cleavage of HC2 could alter the physiological and mechanical properties of the extracellular matrix and contribute to the progression of arthritis.


Subject(s)
ADAMTS5 Protein/metabolism , Alpha-Globulins/metabolism , Arthritis/enzymology , Synovial Fluid/enzymology , ADAMTS5 Protein/genetics , Alpha-Globulins/chemistry , Alpha-Globulins/genetics , Amino Acid Motifs , Arthritis/genetics , Arthritis/metabolism , Extracellular Matrix/enzymology , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Humans , Hyaluronic Acid/metabolism , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase 7/genetics , Matrix Metalloproteinase 7/metabolism , Synovial Fluid/metabolism
5.
Bioconjug Chem ; 30(5): 1356-1370, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30966735

ABSTRACT

This work discloses the first examples of antibody-drug conjugates (ADCs) that are constructed from linker-drugs bearing dimeric seco-CBI payloads (duocarmycin analogs). Several homogeneous, CD22-targeting THIOMAB antibody-drug conjugates (TDCs) containing the dimeric seco-CBI entities are shown to be highly efficacious in the WSU-DLCL2 and BJAB mouse xenograft models. Surprisingly, the seco-CBI-containing conjugates are also observed to undergo significant biotransformation in vivo in mice, rats, and monkeys and thereby form 1:1 adducts with the Alpha-1-Microglobulin (A1M) plasma protein from these species. Variation of both the payload mAb attachment site and length of the linker-drug is shown to alter the rates of adduct formation. Subsequent experiments demonstrated that adduct formation attenuates the in vitro antiproliferation activity of the affected seco-CBI-dimer TDCs, but does not significantly impact the in vivo efficacy of the conjugates. In vitro assays employing phosphatase-treated whole blood suggest that A1M adduct formation is likely to occur if the seco-CBI-dimer TDCs are administered to humans. Importantly, protein adduct formation leads to the underestimation of total antibody (Tab) concentrations using an ELISA assay but does not affect Tab values determined via an orthogonal LC-MS/MS method. Several recommendations regarding bioanalysis of future in vivo studies involving related seco-CBI-containing ADCs are provided based on these collective findings.


Subject(s)
Alpha-Globulins/chemistry , Antineoplastic Agents/pharmacology , Immunoconjugates/pharmacology , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dimerization , Haplorhini , Humans , Immunoconjugates/chemistry , Mice , Rats , Xenograft Model Antitumor Assays
6.
Genes Genet Syst ; 92(5): 251-256, 2018 Apr 10.
Article in English | MEDLINE | ID: mdl-29199250

ABSTRACT

A full-length cDNA clone encoding grass carp (Ctenopharyngodon idellus) α1-microglobulin/bikunin precursor (Ci-AMBP) was isolated by subtracted differential hybridization screening from a liver cDNA library. The deduced amino acid sequence shared approximately 50% sequence identity with its mammalian counterparts, but more than 90% identity with another fish species. AMBPs are the precursors of the plasma glycoproteins α1-microglobulin (α1m) and bikunin. Both peptide structures and their chromosomal organization were well conserved in Ci-AMBP. The α1m and bikunin polypeptides are separated by the typical tetrapeptide R-A-R-R that provides an endoproteolytic cleavage site for maturation. The genetic organization of domains and functional motifs indicated that Ci-AMBP is a typical member of the lipocalin and Kunitz-type protease inhibitor superfamilies. Expression of the Ci-AMBP gene in different tissues/organs was evaluated using semi-quantitative RT-PCR and, in contrast to the restricted expression in other species, transcripts were detected in a wide range of tissues. The most abundant expression occurred in the secretory organs, which supports the roles of α1m and bikunin in the immune response to diseases and in the stress response.


Subject(s)
Alpha-Globulins/genetics , Alpha-Globulins/metabolism , Carps/metabolism , Alpha-Globulins/chemistry , Amino Acid Sequence , Animals , Carps/genetics , Cloning, Molecular , Conserved Sequence , Fish Proteins/chemistry , Fish Proteins/genetics , Fish Proteins/metabolism , Liver/metabolism , Multigene Family , Protein Conformation , Tissue Distribution
7.
Int J Biol Macromol ; 108: 936-941, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29104054

ABSTRACT

Gold nanoparticles are new kinds of nanomaterials. Their large surface-to-volume ratio, stability, excellent biocompatibility, low toxicity and functionality make them very attractive for biomedical applications. Therefore we have analyzed how dendronized gold nanoparticles interact with human alpha-1-microglobulin. This is a glycoprotein of ∼30kDa present in blood plasma and some tissues of the human body. Comparing 3 nanoparticles with different dendronization, we conclude that the effect of a nanoparticle on the structure of alpha-1-microglobulin significantly decreased with second and third generations dendrons as a result of less exposure of the metal cores in the nanoparticles. These interactions indicate weak changes in the immunochemical properties of the protein, whereas the dendron coating had no effect. Thus, dendronization of gold nanoparticles helps to modify their binding properties by shielding them from interactions with plasma proteins.


Subject(s)
Alpha-Globulins/chemistry , Dendrimers/chemistry , Gold/chemistry , Metal Nanoparticles/chemistry , Humans , Models, Molecular , Particle Size , Protein Conformation
8.
J Biol Chem ; 292(51): 20845-20858, 2017 12 22.
Article in English | MEDLINE | ID: mdl-29122888

ABSTRACT

Exposure to pollutants, such as ozone, exacerbates airway inflammation and hyperresponsiveness (AHR). TNF-stimulated gene 6 (TSG-6) is required to transfer inter-α-inhibitor heavy chains (HC) to hyaluronan (HA), facilitating HA receptor binding. TSG-6 is necessary for AHR in allergic asthma, because it facilitates the development of a pathological HA-HC matrix. However, the role of TSG-6 in acute airway inflammation is not well understood. Here, we hypothesized that TSG-6 is essential for the development of HA- and ozone-induced AHR. TSG-6-/- and TSG-6+/+ mice were exposed to ozone or short-fragment HA (sHA), and AHR was assayed via flexiVent. The AHR response to sHA was evaluated in the isolated tracheal ring assay in tracheal rings from TSG-6-/- or TSG-6+/+, with or without the addition of exogenous TSG-6, and with or without inhibitors of Rho-associated, coiled-coil-containing protein kinase (ROCK), ERK, or PI3K. Smooth-muscle cells from mouse tracheas were assayed in vitro for signaling pathways. We found that TSG-6 deficiency protects against AHR after ozone (in vivo) or sHA (in vitro and in vivo) exposure. Moreover, TSG-6-/- tracheal ring non-responsiveness to sHA was reversed by exogenous TSG-6 addition. sHA rapidly activated RhoA, ERK, and Akt in airway smooth-muscle cells, but only in the presence of TSG-6. Inhibition of ROCK, ERK, or PI3K/Akt blocked sHA/TSG-6-mediated AHR. In conclusion, TSG-6 is necessary for AHR in response to ozone or sHA, in part because it facilitates rapid formation of HA-HC complexes. The sHA/TSG-6 effect is mediated by RhoA, ERK, and PI3K/Akt signaling.


Subject(s)
Alpha-Globulins/metabolism , Cell Adhesion Molecules/metabolism , Hyaluronic Acid/metabolism , Respiratory Hypersensitivity/etiology , Respiratory Hypersensitivity/metabolism , Air Pollutants/toxicity , Alpha-Globulins/chemistry , Animals , Cell Adhesion Molecules/deficiency , Cell Adhesion Molecules/genetics , Disease Models, Animal , Hyaluronic Acid/chemistry , In Vitro Techniques , Macromolecular Substances/chemistry , Macromolecular Substances/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Models, Biological , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Ozone/toxicity , Signal Transduction/drug effects , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein
9.
Eur J Pharmacol ; 802: 60-68, 2017 May 05.
Article in English | MEDLINE | ID: mdl-28249709

ABSTRACT

Severe sepsis is a complex, multifactorial, and rapidly progressing disease characterized by excessive inflammation and coagulation following bacterial infection. To simultaneously suppress pro-inflammatory and pro-coagulant responses, we genetically engineered a novel fusion protein (MR1007) consisting of an anti-CD14 antibody and the modified second domain of bikunin, and evaluated the potential of MR1007 as an anti-sepsis agent. Suppressive effects of MR1007 on lipopolysaccharide (LPS)-induced inflammatory responses were assessed using peripheral blood mononuclear cells or endothelial cells. Its inhibitory activity against the coagulation factor XIa was assessed using a purified enzyme and a chromogenic substrate. Anticoagulant activity was assessed using human or rabbit plasma. Anti-inflammatory and anti-coagulant effects and/or survival benefits were evaluated in an endotoxemia model and a cecal ligation and puncture model. MR1007 inhibited LPS-induced cytokine production in peripheral blood mononuclear cells and endothelial cells, inhibited factor XIa, and exhibited anticoagulant activity. In an endotoxemia model, 0.3-3mg/kg MR1007 suppressed pro-inflammatory and pro-coagulant responses in a dose-dependent manner; at a dose of 3mg/kg, the protein improved survival even when administered 8h after the LPS injection. In addition, 10mg/kg MR1007 administered 2h post cecal ligation and puncture improved survival. However, MR1007 administered at doses up to 30mg/kg did not increase ear bleeding time or bacterial counts in the cecal ligation and puncture model. Thus, simultaneous targeting of CD14 and factor XIa improves survival in the rabbit endotoxemia and sepsis models and represents a promising approach for the treatment of severe sepsis.


Subject(s)
Alpha-Globulins/chemistry , Antibodies, Monoclonal/pharmacology , Endotoxemia/drug therapy , Factor XIa/antagonists & inhibitors , Lipopolysaccharide Receptors/immunology , Molecular Targeted Therapy , Recombinant Fusion Proteins/pharmacology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Anticoagulants/immunology , Anticoagulants/pharmacology , Anticoagulants/therapeutic use , Bleeding Time , Disease Models, Animal , E-Selectin/metabolism , Endotoxemia/metabolism , Gene Expression Regulation/drug effects , Humans , Interleukin-6/biosynthesis , Protein Domains , Rabbits , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/therapeutic use , Survival Analysis
10.
J Am Soc Mass Spectrom ; 28(2): 229-241, 2017 02.
Article in English | MEDLINE | ID: mdl-27873218

ABSTRACT

Purification and liquid chromatography-tandem mass spectrometry (LC-MS/MS) characterization of glycopeptides, originating from protease digests of glycoproteins, enables site-specific analysis of protein N- and O-glycosylations. We have described a protocol to enrich, hydrolyze by chondroitinase ABC, and characterize chondroitin sulfate-containing glycopeptides (CS-glycopeptides) using positive mode LC-MS/MS. The CS-glycopeptides, originating from the Bikunin proteoglycan of human urine samples, had ΔHexAGalNAcGlcAGalGalXyl-O-Ser hexasaccharide structure and were further substituted with 0-3 sulfate and 0-1 phosphate groups. However, it was not possible to exactly pinpoint sulfate attachment residues, for protonated precursors, due to extensive fragmentation of sulfate groups using high-energy collision induced dissociation (HCD). To circumvent the well-recognized sulfate instability, we now introduced Na+ ions to form sodiated precursors, which protected sulfate groups from decomposition and facilitated the assignment of sulfate modifications. Sulfate groups were pinpointed to both Gal residues and to the GalNAc of the hexasaccharide structure. The intensities of protonated and sodiated saccharide oxonium ions were very prominent in the HCD-MS2 spectra, which provided complementary structural analysis of sulfate substituents of CS-glycopeptides. We have demonstrated a considerable heterogeneity of the bikunin CS linkage region. The realization of these structural variants should be beneficial in studies aimed at investigating the importance of the CS linkage region with regards to the biosynthesis of CS and potential interactions to CS binding proteins. Also, the combined use of protonated and sodiated precursors for positive mode HCD fragmentation analysis will likely become useful for additional classes of sulfated glycopeptides. Graphical Abstract ᅟ.


Subject(s)
Chondroitin Sulfates/chemistry , Chromatography, Liquid/methods , Glycopeptides/chemistry , Polysaccharides/chemistry , Tandem Mass Spectrometry/methods , Acetylgalactosamine/chemistry , Alpha-Globulins/chemistry , Alpha-Globulins/urine , Carbohydrate Sequence , Humans , N-Acetylneuraminic Acid/chemistry , Phosphorylation , Polysaccharides/analysis , Sodium/chemistry
11.
J Biol Chem ; 291(9): 4658-70, 2016 Feb 26.
Article in English | MEDLINE | ID: mdl-26728454

ABSTRACT

Inter-α-inhibitor is a proteoglycan of unique structure. The protein consists of three subunits, heavy chain 1, heavy chain 2, and bikunin covalently joined by a chondroitin sulfate chain originating at Ser-10 of bikunin. Inter-α-inhibitor interacts with an inflammation-associated protein, tumor necrosis factor-inducible gene 6 protein, in the extracellular matrix. This interaction leads to transfer of the heavy chains from the chondroitin sulfate of inter-α-inhibitor to hyaluronan and consequently to matrix stabilization. Divalent cations and heavy chain 2 are essential co-factors in this transfer reaction. In the present study, we have investigated how divalent cations in concert with the chondroitin sulfate chain influence the structure and stability of inter-α-inhibitor. The results showed that Mg(2+) or Mn(2+), but not Ca(2+), induced a conformational change in inter-α-inhibitor as evidenced by a decrease in the Stokes radius and a bikunin chondroitin sulfate-dependent increase of the thermodynamic stability. This structure was shown to be essential for the ability of inter-α-inhibitor to participate in extracellular matrix stabilization. In addition, the data revealed that bikunin was positioned adjacent to both heavy chains and that the two heavy chains also were in close proximity. The chondroitin sulfate chain interacted with all protein components and inter-α-inhibitor dissociated when it was degraded. Conventional purification protocols result in the removal of the Mg(2+) found in plasma and because divalent cations influence the conformation and affect function it is important to consider this when characterizing the biological activity of inter-α-inhibitor.


Subject(s)
Alpha-Globulins/chemistry , Chondroitin Sulfates/chemistry , Magnesium/chemistry , Manganese/chemistry , Models, Molecular , Proteoglycans/chemistry , Alpha-Globulins/isolation & purification , Alpha-Globulins/metabolism , Binding Sites , Chondroitin Sulfates/metabolism , Cross-Linking Reagents/chemistry , Hot Temperature/adverse effects , Humans , Ligands , Magnesium/metabolism , Manganese/metabolism , Molecular Conformation , Protein Conformation , Protein Footprinting , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Stability , Protein Subunits/chemistry , Protein Subunits/isolation & purification , Protein Subunits/metabolism , Protein Unfolding , Proteoglycans/metabolism
12.
Biochim Biophys Acta ; 1864(1): 29-41, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26497278

ABSTRACT

BACKGROUND: α1-Microglobulin (A1M) is a reductase and radical scavenger involved in physiological protection against oxidative damage. These functions were previously shown to be dependent upon cysteinyl-, C34, and lysyl side-chains, K(92, 118,130). A1M binds heme and the crystal structure suggests that C34 and H123 participate in a heme binding site. We have investigated the involvement of these five residues in the interactions with heme. METHODS: Four A1M-variants were expressed: with cysteine to serine substitution in position 34, lysine to threonine substitutions in positions (92, 118, 130), histidine to serine substitution in position 123 and a wt without mutations. Heme binding was investigated by tryptophan fluorescence quenching, UV-Vis spectrophotometry, circular dichroism, SPR, electrophoretic migration shift, gel filtration, catalase-like activity and molecular simulation. RESULTS: All A1M-variants bound to heme. Mutations in C34, H123 or K(92, 118, 130) resulted in significant absorbance changes, CD spectral changes, and catalase-like activity, suggesting involvement of these side-groups in coordination of the heme-iron. Molecular simulation support a model with two heme-binding sites in A1M involving the mutated residues. Binding of the first heme induces allosteric stabilization of the structure predisposing for a better fit of the second heme. CONCLUSIONS: The results suggest that one heme-binding site is located in the lipocalin pocket and a second binding site between loops 1 and 4. Reactions with the hemes involve the side-groups of C34, K(92, 118, 130) and H123. GENERAL SIGNIFICANCE: The model provides a structural basis for the functional activities of A1M: heme binding activity of A1M.


Subject(s)
Alpha-Globulins/chemistry , Heme/chemistry , Molecular Dynamics Simulation , Protein Structure, Tertiary , Alpha-Globulins/genetics , Alpha-Globulins/metabolism , Binding Sites/genetics , Blotting, Western , Circular Dichroism , Heme/metabolism , Humans , Mutagenesis, Site-Directed/methods , Mutation , Oxidation-Reduction , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Spectrometry, Fluorescence , Surface Plasmon Resonance
13.
Mol Cell Proteomics ; 14(12): 3118-31, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26407992

ABSTRACT

The inter-α-trypsin inhibitor complex is a macromolecular arrangement of structurally related heavy chain proteins covalently cross-linked to the chondroitin sulfate (CS) chain of the proteoglycan bikunin. The inter-α-trypsin inhibitor complex is abundant in plasma and associated with inflammation, kidney diseases, cancer and diabetes. Bikunin is modified at Ser-10 by a single low-sulfated CS chain of 23-55 monosaccharides with 4-9 sulfate groups. The innermost four monosaccharides (GlcAß3Galß3Galß4Xylß-O-) compose the linkage region, believed to be uniform with a 4-O-sulfation to the outer Gal. The cross-linkage region of the bikunin CS chain is located in the nonsulfated nonreducing end, (GalNAcß4GlcAß3)(n), to which heavy chains (H1-H3) may be bound in GalNAc to Asp ester linkages. In this study we employed a glycoproteomics protocol to enrich and analyze light and heavy chain linkage and cross-linkage region CS glycopeptides derived from the IαI complex of human plasma, urine and cerebrospinal fluid samples. The samples were trypsinized, enriched by strong anion exchange chromatography, partially depolymerized with chondroitinase ABC and analyzed by LC-MS/MS using higher-energy collisional dissociation. The analyses demonstrated that the CS linkage region of bikunin is highly heterogeneous. In addition to sulfation of the Gal residue, Xyl phosphorylation was observed although exclusively in urinary samples. We also identified novel Neu5Ac and Fuc modifications of the linkage region as well as the presence of mono- and disialylated core 1 O-linked glycans on Thr-17. Heavy chains H1 and H2 were identified cross-linked to GalNAc residues one or two GlcA residues apart and H1 was found linked to either the terminal or subterminal GalNAc residues. The fragmentation behavior of CS glycopeptides under variable higher-energy collisional dissociation conditions displays an energy dependence that may be used to obtain complementary structural details. Finally, we show that the analysis of sodium adducts provides confirmatory information about the positions of glycan substituents.


Subject(s)
Alpha-Globulins/isolation & purification , Chondroitin Sulfates/chemistry , Proteomics/methods , Alpha-Globulins/cerebrospinal fluid , Alpha-Globulins/chemistry , Alpha-Globulins/urine , Chromatography, Liquid/methods , Galactose/chemistry , Glycopeptides/chemistry , Glycopeptides/isolation & purification , Humans , Male , Tandem Mass Spectrometry/methods
14.
Biomacromolecules ; 16(3): 951-61, 2015 Mar 09.
Article in English | MEDLINE | ID: mdl-25645947

ABSTRACT

Proteoglycan spatiotemporal organization underpins extracellular matrix biology, but atomic scale glimpses of this microarchitecture are obscured by glycosaminoglycan size and complexity. To overcome this, multimicrosecond aqueous simulations of chondroitin and dermatan sulfates were abstracted into a prior coarse-grained model, which was extended to heterogeneous glycosaminoglycans and small leucine-rich proteoglycans. Exploration of relationships between sequence and shape led to hypotheses that proteoglycan size is dependent on glycosaminoglycan unit composition but independent of sequence permutation. Uronic acid conformational equilibria were modulated by adjacent hexosamine sulfonation and iduronic acid increased glycosaminoglycan chain volume and rigidity, while glucuronic acid imparted chain plasticity. Consequently, block copolymeric glycosaminoglycans contained microarchitectures capable of multivalent binding to growth factors and collagen, with potential for interactional synergy at greater chain number. The described atomic scale views of proteoglycans and heterogeneous glycosaminoglycans provide structural routes to understanding their fundamental signaling and mechanical biological roles and development of new biomaterials.


Subject(s)
Alpha-Globulins/chemistry , Chondroitin Sulfates/chemistry , Decorin/chemistry , Dermatan Sulfate/chemistry , Animals , Carbohydrate Conformation , Cartilage/chemistry , Cattle , Humans , Intestines/chemistry , Molecular Dynamics Simulation , Protein Conformation , Sharks , Sus scrofa , Trachea/chemistry
15.
Mol Cell Proteomics ; 14(1): 41-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25326458

ABSTRACT

Vertebrates produce various chondroitin sulfate proteoglycans (CSPGs) that are important structural components of cartilage and other connective tissues. CSPGs also contribute to the regulation of more specialized processes such as neurogenesis and angiogenesis. Although many aspects of CSPGs have been studied extensively, little is known of where the CS chains are attached on the core proteins and so far, only a limited number of CSPGs have been identified. Obtaining global information on glycan structures and attachment sites would contribute to our understanding of the complex proteoglycan structures and may also assist in assigning CSPG specific functions. In the present work, we have developed a glycoproteomics approach that characterizes CS linkage regions, attachment sites, and identities of core proteins. CSPGs were enriched from human urine and cerebrospinal fluid samples by strong-anion-exchange chromatography, digested with chondroitinase ABC, a specific CS-lyase used to reduce the CS chain lengths and subsequently analyzed by nLC-MS/MS with a novel glycopeptide search algorithm. The protocol enabled the identification of 13 novel CSPGs, in addition to 13 previously established CSPGs, demonstrating that this approach can be routinely used to characterize CSPGs in complex human samples. Surprisingly, five of the identified CSPGs are traditionally defined as prohormones (cholecystokinin, chromogranin A, neuropeptide W, secretogranin-1, and secretogranin-3), typically stored and secreted from granules of endocrine cells. We hypothesized that the CS side chain may influence the assembly and structural organization of secretory granules and applied surface plasmon resonance spectroscopy to show that CS actually promotes the assembly of chromogranin A core proteins in vitro. This activity required mild acidic pH and suggests that the CS-side chains may also influence the self-assembly of chromogranin A in vivo giving a possible explanation to previous observations that chromogranin A has an inherent property to assemble in the acidic milieu of secretory granules.


Subject(s)
Alpha-Globulins , Chondroitin Sulfate Proteoglycans , Glycopeptides , Alpha-Globulins/cerebrospinal fluid , Alpha-Globulins/chemistry , Alpha-Globulins/metabolism , Alpha-Globulins/urine , Cholecystokinin/analysis , Chondroitin Sulfate Proteoglycans/cerebrospinal fluid , Chondroitin Sulfate Proteoglycans/chemistry , Chondroitin Sulfate Proteoglycans/metabolism , Chondroitin Sulfate Proteoglycans/urine , Chromogranin A/analysis , Chromogranin B/analysis , Chromogranins/analysis , Glycopeptides/cerebrospinal fluid , Glycopeptides/chemistry , Glycopeptides/metabolism , Glycopeptides/urine , Humans , Male , Neuropeptides/analysis
16.
Indian J Exp Biol ; 53(12): 771-8, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26742321

ABSTRACT

In mammals, a low molecular mass protein (17-20 KDa) reported from the pheromone sources such as urine, saliva, glandular secretion, etc., as ligand-carrier (pheromone carrier) has been associated with chemo-communication. Since the preorbital gland post is one of the major pheromone sources in Indian Blackbuck, an endangered species, we assumed that it possibly contains low molecular mass protein for chemical communication. Hence, we investigated the preorbital gland post in territorial and non-territorial male blackbucks for such low molecular mass proteins adopting SDS-PAGE and LC-MS/MS analysis. The total content of protein was higher in the post of territorial males than non-territorial males of adult and sub-adult. In fact, the protein profiles such as 17, 21, 25, 42 and 61 kDa were noted in the gland secretion of territorial and non-territorial males. The intensity of the 17 kDa protein band was higher in territorial males than non-territorial males. In-gel trypsin digestion of the 17 kDa band was processed and subjected to LC-MS/MS and SEQUEST analyses. The results of LC-MS/MS and SEQUEST search showed the presence of α(2u)-globulin in the 17 kDa band. In addition, the identified α(2u)-globulin sequence possessed GDW residues, which are the characteristic signature for lipocalin family. Since the α(2u)-globulin has been reported from the pheromone-carrying proteins in some mammals, this protein may carry the volatiles (pheromone compounds) in male Blackbucks preorbital gland to evoke the scent marking for maintaining territoriality (home range) and attraction towards female, through the secretion of glandular protein.


Subject(s)
Alpha-Globulins/metabolism , Antelopes/metabolism , Carrier Proteins/metabolism , Endangered Species , Exocrine Glands/metabolism , Pheromones/metabolism , Alpha-Globulins/chemistry , Amino Acid Sequence , Animal Communication , Animals , Antelopes/genetics , Antelopes/psychology , Carrier Proteins/chemistry , Chromatography, Liquid , Electrophoresis, Polyacrylamide Gel , Male , Molecular Sequence Data , Molecular Weight , Proteomics/methods , Tandem Mass Spectrometry , Territoriality
17.
Biosci Biotechnol Biochem ; 79(4): 566-73, 2015.
Article in English | MEDLINE | ID: mdl-25522807

ABSTRACT

Cereal prolamins, which are alcohol-soluble seed storage proteins, can induce ER-derived protein bodies (PBs) in heterologous tissue. Like maize and wheat prolamins, rice prolamins can form ER-derived PBs, but the region of mature polypeptides that is essential for PB formation has not been identified. In this study, we examined the formation mechanisms of ER-derived PB-like structures by expressing rice 13 kDa prolamin-deletion mutants fused to green fluorescent protein (GFP) in heterologous tissues such as yeast. The 13 kDa prolamin-GFP fusion protein was stably accumulated in transgenic yeast and formed an ER-derived PB-like structure. In contrast, rice α-globulin-GFP fusion protein was transported to vacuoles. In addition, the middle and COOH-terminal regions of 13 kDa prolamin formed ER-derived PB-like structures, whereas the NH2-terminal region of 13 kDa prolamin did not form such structures. These results suggest that the middle and COOH-terminal regions of 13 kDa prolamin can be retained and thus can induce ER-derived PB in yeast.


Subject(s)
Oryza/genetics , Prolamins/chemistry , Recombinant Fusion Proteins/chemistry , Seeds/genetics , Alpha-Globulins/chemistry , Alpha-Globulins/genetics , Alpha-Globulins/metabolism , Endoplasmic Reticulum/metabolism , Gene Expression , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Oryza/metabolism , Plasmids/chemistry , Plasmids/metabolism , Prolamins/genetics , Prolamins/metabolism , Protein Transport , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Seeds/metabolism , Vacuoles/metabolism
18.
J Biol Chem ; 289(47): 32783-97, 2014 Nov 21.
Article in English | MEDLINE | ID: mdl-25301953

ABSTRACT

Mesotrypsin is an isoform of trypsin that is uniquely resistant to polypeptide trypsin inhibitors and can cleave some inhibitors rapidly. Previous studies have shown that the amyloid precursor protein Kunitz protease inhibitor domain (APPI) is a specific substrate of mesotrypsin and that stabilization of the APPI cleavage site in a canonical conformation contributes to recognition by mesotrypsin. We hypothesized that other proteins possessing potential cleavage sites stabilized in a similar conformation might also be mesotrypsin substrates. Here we evaluated a series of candidate substrates, including human Kunitz protease inhibitor domains from amyloid precursor-like protein 2 (APLP2), bikunin, hepatocyte growth factor activator inhibitor type 2 (HAI2), tissue factor pathway inhibitor-1 (TFPI1), and tissue factor pathway inhibitor-2 (TFPI2), as well as E-selectin, an unrelated protein possessing a potential cleavage site displaying canonical conformation. We find that Kunitz domains within APLP2, bikunin, and HAI2 are cleaved by mesotrypsin with kinetic profiles of specific substrates. TFPI1 and TFPI2 Kunitz domains are cleaved less efficiently by mesotrypsin, and E-selectin is not cleaved at the anticipated site. Cocrystal structures of mesotrypsin with HAI2 and bikunin Kunitz domains reveal the mode of mesotrypsin interaction with its canonical substrates. Our data suggest that major determinants of mesotrypsin substrate specificity include sequence preferences at the P1 and P'2 positions along with conformational stabilization of the cleavage site in the canonical conformation. Mesotrypsin up-regulation has been implicated previously in cancer progression, and proteolytic clearance of Kunitz protease inhibitors offers potential mechanisms by which mesotrypsin may mediate pathological effects in cancer.


Subject(s)
Protease Inhibitors/chemistry , Protein Conformation , Protein Structure, Tertiary , Trypsin/chemistry , Alpha-Globulins/chemistry , Alpha-Globulins/genetics , Alpha-Globulins/metabolism , Amino Acid Sequence , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Aprotinin/chemistry , Aprotinin/genetics , Aprotinin/metabolism , Binding Sites/genetics , Crystallography, X-Ray , E-Selectin/chemistry , E-Selectin/genetics , E-Selectin/metabolism , Glycoproteins/chemistry , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Kinetics , Lipoproteins/chemistry , Lipoproteins/genetics , Lipoproteins/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Models, Molecular , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Protease Inhibitors/metabolism , Protein Binding , Substrate Specificity , Trypsin/genetics , Trypsin/metabolism
19.
J Biol Chem ; 289(44): 30481-30498, 2014 Oct 31.
Article in English | MEDLINE | ID: mdl-25190808

ABSTRACT

Mammalian oocytes are surrounded by a highly hydrated hyaluronan (HA)-rich extracellular matrix with embedded cumulus cells, forming the cumulus cell·oocyte complex (COC) matrix. The correct assembly, stability, and mechanical properties of this matrix, which are crucial for successful ovulation, transport of the COC to the oviduct, and its fertilization, depend on the interaction between HA and specific HA-organizing proteins. Although the proteins inter-α-inhibitor (IαI), pentraxin 3 (PTX3), and TNF-stimulated gene-6 (TSG-6) have been identified as being critical for COC matrix formation, its supramolecular organization and the molecular mechanism of COC matrix stabilization remain unknown. Here we used films of end-grafted HA as a model system to investigate the molecular interactions involved in the formation and stabilization of HA matrices containing TSG-6, IαI, and PTX3. We found that PTX3 binds neither to HA alone nor to HA films containing TSG-6. This long pentraxin also failed to bind to products of the interaction between IαI, TSG-6, and HA, among which are the covalent heavy chain (HC)·HA and HC·TSG-6 complexes, despite the fact that both IαI and TSG-6 are ligands of PTX3. Interestingly, prior encounter with IαI was required for effective incorporation of PTX3 into TSG-6-loaded HA films. Moreover, we demonstrated that this ternary protein mixture made of IαI, PTX3, and TSG-6 is sufficient to promote formation of a stable (i.e. cross-linked) yet highly hydrated HA matrix. We propose that this mechanism is essential for correct assembly of the COC matrix and may also have general implications in other inflammatory processes that are associated with HA cross-linking.


Subject(s)
C-Reactive Protein/chemistry , Extracellular Matrix/physiology , Hyaluronic Acid/chemistry , Serum Amyloid P-Component/chemistry , Alpha-Globulins/chemistry , Animals , Cell Adhesion Molecules/chemistry , Cell Line , Drosophila melanogaster , Extracellular Matrix/chemistry , Female , Humans , Ovarian Follicle/metabolism , Protein Binding
20.
J Biol Chem ; 289(42): 29171-9, 2014 Oct 17.
Article in English | MEDLINE | ID: mdl-25135638

ABSTRACT

We have recently demonstrated that the transfer of heavy chains (HCs) from inter-α-inhibitor, via the enzyme TSG-6 (tumor necrosis factor-stimulated gene 6), to hyaluronan (HA) oligosaccharides is an irreversible event in which subsequent swapping of HCs between HA molecules does not occur. We now describe our results of HC transfer experiments to chondroitin sulfate A, chemically desulfated chondroitin, chemoenzymatically synthesized chondroitin, unsulfated heparosan, heparan sulfate, and alginate. Of these potential HC acceptors, only chemically desulfated chondroitin and chemoenzymatically synthesized chondroitin were HC acceptors. The kinetics of HC transfer to chondroitin was similar to HA. At earlier time points, HCs were more widely distributed among the different sizes of chondroitin chains. As time progressed, the HCs migrated to lower molecular weight chains of chondroitin. Our interpretation is that TSG-6 swaps the HCs from the larger, reversible sites on chondroitin chains, which function as HC acceptors, onto smaller chondroitin chains, which function as irreversible HC acceptors. HCs transferred to smaller chondroitin chains were unable to be swapped off the smaller chondroitin chains and transferred to HA. HCs transferred to high molecular weight HA were unable to be swapped onto chondroitin. We also present data that although chondroitin was a HC acceptor, HA was the preferred acceptor when chondroitin and HA were in the same reaction mixture.


Subject(s)
Chondroitin/chemistry , Hyaluronic Acid/chemistry , Oligosaccharides/chemistry , Alginates/chemistry , Alpha-Globulins/chemistry , Cell Adhesion Molecules/chemistry , Chondroitin Sulfates/chemistry , Disaccharides/chemistry , Glucuronic Acid/chemistry , Heparitin Sulfate/chemistry , Hexuronic Acids/chemistry , Humans , Kinetics , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...