Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
1.
Circ Res ; 132(9): e151-e168, 2023 04 28.
Article in English | MEDLINE | ID: mdl-37021588

ABSTRACT

BACKGROUND: Neutrophil migration is critical to the initiation and resolution of inflammation. Macrophage-1 antigen (Mac-1; CD11b/CD18, αMß2) is a leukocyte integrin essential for firm adhesion to endothelial ICAM-1 (intercellular adhesion molecule 1) and migration of neutrophils in the shear forces of the circulation. PDI (protein disulfide isomerase) has been reported to influence neutrophil adhesion and migration. We aimed to elucidate the molecular mechanism of PDI control of Mac-1 affinity for ICAM-1 during neutrophil migration under fluid shear. METHODS: Neutrophils isolated from whole blood were perfused over microfluidic chips coated with ICAM-1. Colocalization of Mac-1 and PDI on neutrophils was visualized by fluorescently labeled antibodies and confocal microscopy. The redox state of Mac-1 disulfide bonds was mapped by differential cysteine alkylation and mass spectrometry. Wild-type or disulfide mutant Mac-1 was expressed recombinantly in Baby Hamster Kidney cells to measure ligand affinity. Mac-1 conformations were measured by conformation-specific antibodies and molecular dynamics simulations. Neutrophils crawling on immobilized ICAM-1 were measured in presence of oxidized or reduced PDI, and the effect of PDI inhibition using isoquercetin on neutrophil crawling on inflamed endothelial cells was examined. Migration indices in the X- and Y-direction were determined and the crawling speed was calculated. RESULTS: PDI colocalized with high-affinity Mac-1 at the trailing edge of stimulated neutrophils when crawling on ICAM-1 under fluid shear. PDI cleaved 2 allosteric disulfide bonds, C169-C176 and C224-C264, in the ßI domain of the ß2 subunit, and cleavage of the C224-C264 disulfide bond selectively controls Mac-1 disengagement from ICAM-1 under fluid shear. Molecular dynamics simulations and conformation-specific antibodies reveal that cleavage of the C224-C264 bond induces conformational change and mechanical stress in the ßI domain. This allosterically alters the exposure of an αI domain epitope associated with a shift of Mac-1 to a lower-affinity state. These molecular events promote neutrophil motility in the direction of flow at high shear stress. Inhibition of PDI by isoquercetin reduces neutrophil migration in the direction of flow on endothelial cells during inflammation. CONCLUSIONS: Shear-dependent PDI cleavage of the neutrophil Mac-1 C224-C264 disulfide bond triggers Mac-1 de-adherence from ICAM-1 at the trailing edge of the cell and enables directional movement of neutrophils during inflammation.


Subject(s)
Intercellular Adhesion Molecule-1 , Macrophage-1 Antigen , Humans , Macrophage-1 Antigen/physiology , Cell Adhesion/physiology , Endothelial Cells , Inflammation , Cell Movement/physiology , Neutrophils
2.
Int J Cancer ; 152(7): 1399-1413, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36346110

ABSTRACT

The mitochondrion is a gatekeeper of apoptotic processes, and mediates drug resistance to several chemotherapy agents used to treat cancer. Neuroblastoma is a common solid cancer in young children with poor clinical outcomes following conventional chemotherapy. We sought druggable mitochondrial protein targets in neuroblastoma cells. Among mitochondria-associated gene targets, we found that high expression of the mitochondrial adenine nucleotide translocase 2 (SLC25A5/ANT2), was a strong predictor of poor neuroblastoma patient prognosis and contributed to a more malignant phenotype in pre-clinical models. Inhibiting this transporter with PENAO reduced cell viability in a panel of neuroblastoma cell lines in a TP53-status-dependant manner. We identified the histone deacetylase inhibitor, suberanilohydroxamic acid (SAHA), as the most effective drug in clinical use against mutant TP53 neuroblastoma cells. SAHA and PENAO synergistically reduced cell viability, and induced apoptosis, in neuroblastoma cells independent of TP53-status. The SAHA and PENAO drug combination significantly delayed tumour progression in pre-clinical neuroblastoma mouse models, suggesting that these clinically advanced inhibitors may be effective in treating the disease.


Subject(s)
Adenine Nucleotide Translocator 2 , Antineoplastic Agents , Histone Deacetylase Inhibitors , Hydroxamic Acids , Neuroblastoma , Animals , Mice , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Hydroxamic Acids/therapeutic use , Mitochondria/metabolism , Neuroblastoma/drug therapy , Vorinostat/pharmacology , Adenine Nucleotide Translocator 2/antagonists & inhibitors
3.
Blood ; 138(15): 1359-1372, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34375384

ABSTRACT

The αIIbß3 integrin receptor coordinates platelet adhesion, activation, and mechanosensing in thrombosis and hemostasis. Using differential cysteine alkylation and mass spectrometry, we have identified a disulfide bond in the αIIb subunit linking cysteines 490 and 545 that is missing in ∼1 in 3 integrin molecules on the resting and activated human platelet surface. This alternate covalent form of αIIbß3 is predetermined as it is also produced by human megakaryoblasts and baby hamster kidney fibroblasts transfected with recombinant integrin. From coimmunoprecipitation experiments, the alternate form selectively partitions into focal adhesions on the activated platelet surface. Its function was evaluated in baby hamster kidney fibroblast cells expressing a mutant integrin with an ablated C490-C545 disulfide bond. The disulfide mutant integrin has functional outside-in signaling but extended residency time in focal adhesions due to a reduced rate of clathrin-mediated integrin internalization and recycling, which is associated with enhanced affinity of the αIIb subunit for clathrin adaptor protein 2. Molecular dynamics simulations indicate that the alternate covalent form of αIIb requires higher forces to transition from bent to open conformational states that is in accordance with reduced affinity for fibrinogen and activation by manganese ions. These findings indicate that the αIIbß3 integrin receptor is produced in various covalent forms that have different cell surface distribution and function. The C490, C545 cysteine pair is conserved across all 18 integrin α subunits, and the disulfide bond in the αV and α2 subunits in cultured cells is similarly missing, suggesting that the alternate integrin form and function are also conserved.


Subject(s)
Focal Adhesions/metabolism , Integrin beta3/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Membrane Glycoprotein IIb/metabolism , Animals , Cell Line , Cricetinae , Disulfides/analysis , Focal Adhesions/genetics , Human Umbilical Vein Endothelial Cells , Humans , Integrin beta3/chemistry , Integrin beta3/genetics , Molecular Dynamics Simulation , Mutation , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Platelet Membrane Glycoprotein IIb/chemistry , Platelet Membrane Glycoprotein IIb/genetics
4.
Proc Natl Acad Sci U S A ; 117(39): 24434-24442, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32917816

ABSTRACT

Sphingolipid dysregulation is often associated with insulin resistance, while the enzymes controlling sphingolipid metabolism are emerging as therapeutic targets for improving insulin sensitivity. We report herein that sphingosine kinase 2 (SphK2), a key enzyme in sphingolipid catabolism, plays a critical role in the regulation of hepatic insulin signaling and glucose homeostasis both in vitro and in vivo. Hepatocyte-specific Sphk2 knockout mice exhibit pronounced insulin resistance and glucose intolerance. Likewise, SphK2-deficient hepatocytes are resistant to insulin-induced activation of the phosphoinositide 3-kinase (PI3K)-Akt-FoxO1 pathway and elevated hepatic glucose production. Mechanistically, SphK2 deficiency leads to the accumulation of sphingosine that, in turn, suppresses hepatic insulin signaling by inhibiting PI3K activation in hepatocytes. Either reexpressing functional SphK2 or pharmacologically inhibiting sphingosine production restores insulin sensitivity in SphK2-deficient hepatocytes. In conclusion, the current study provides both experimental findings and mechanistic data showing that SphK2 and sphingosine in the liver are critical regulators of insulin sensitivity and glucose homeostasis.


Subject(s)
Glucose/metabolism , Insulin/metabolism , Liver/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Animals , Cell Line, Tumor , Female , Hepatocytes/enzymology , Hepatocytes/metabolism , Homeostasis , Humans , Liver/enzymology , Male , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Sphingolipids/metabolism
5.
Eur J Nucl Med Mol Imaging ; 49(12): 4037-4047, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35779082

ABSTRACT

PURPOSE: This study assesses human biodistribution, radiation dosimetry, safety and tumour uptake of cell death indicator labelled with 68Ga ([68Ga]Ga-CDI), a novel radiopharmaceutical that can image multiple forms of cell death. METHODS: Five participants with at least one extracranial site of solid malignancy > 2 cm and no active cancer treatment in the 8 weeks prior to the study were enrolled. Participants were administered 205 ± 4.1 MBq (range, 200-211 MBq) of [68Ga]Ga-CDI and 8 serial PET scans acquired: the first commencing immediately and the last 3 h later. Participants were monitored for clinical, laboratory and electrocardiographic side effects and adverse events. Urine and blood radioactivity was measured. Spherical volumes of interest were drawn over tumour, blood pool and organs to determine biodistribution and calculate dosimetry. In one participant, tumour specimens were analysed for cell death using terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL) staining. RESULTS: [68Ga]Ga-CDI is safe and well-tolerated with no side effects or adverse events. [68Ga]Ga-CDI is renally excreted, demonstrates low levels of physiologic uptake in the other organs and has excellent imaging characteristics. The mean effective dose was 2.17E - 02 ± 4.61E - 03 mSv/MBq. It images constitutive tumour cell death and correlates with tumour cell death on histology. CONCLUSION: [68Ga]Ga-CDI is a novel cell death imaging radiopharmaceutical that is safe, has low radiation dosimetry and excellent biodistribution and imaging characteristics. It has potential advantages over previously investigated radiopharmaceuticals for imaging of cell death and has progressed to a proof-of-concept trial. TRIAL REGISTRATION: ACTRN12621000641897 (28/5/2021, retrospectively registered).


Subject(s)
Neoplasms , Radiopharmaceuticals , Cell Death , DNA Nucleotidylexotransferase/metabolism , Electrons , Gallium Radioisotopes , Humans , Neoplasms/diagnostic imaging , Neoplasms/metabolism , Positron Emission Tomography Computed Tomography/methods , Positron-Emission Tomography/adverse effects , Positron-Emission Tomography/methods , Radiometry , Radiopharmaceuticals/adverse effects , Tissue Distribution
6.
Invest New Drugs ; 39(3): 756-763, 2021 06.
Article in English | MEDLINE | ID: mdl-33469723

ABSTRACT

PENAO (4-(N-(S-penicillaminylacetyl)amino)phenylarsonous acid) is a second-generation peptide arsenical that inactivates mitochondria in proliferating tumour cells by covalently reacting with mitochondrial inner-membrane adenine nucleotide transferase. The toxicokinetics of PENAO has been investigated in Sprague-Dawley rats to inform route of administration and dosing for human clinical trials. PENAO was well tolerated at 3.3 mg/kg daily intravenous injections but associated with significant toxicity at 10 mg/kg, primarily in the males. The major target organ for toxic effects was the kidney, with changes observed in tubular dilation, presence of casts, basophilic tubules, lymphoid aggregates and interstitial fibrosis. Kidney function was impaired in males with dose-dependent increase in serum creatinine concentration. The severity of the microscopic lesions was reduced in the females, but not the males, at the completion of the four-week recovery period. The elimination phase half-life of PENAO varied between 0.4 and 1.7 h and volume of distribution ranged from 0.25 to 0.88 L/kg for the different dose groups and treatment days, suggesting that PENAO distributes in the extracellular fluids at the doses tested. The area under the curve and clearance values indicate that male rats had reduced elimination of PENAO compared to females, which may account for the increased toxicity in males. PENAO is significantly better tolerated in rodents than its predecessor, GSAO. As GSAO was generally well tolerated with few side effects in a phase I trial in patients with solid tumours, these findings bode well for the tolerability of intravenous dosing of PENAO in patients.


Subject(s)
Antineoplastic Agents , Arsenicals , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Area Under Curve , Arsenicals/adverse effects , Arsenicals/blood , Arsenicals/pharmacokinetics , Creatinine/blood , Female , Half-Life , Kidney/drug effects , Kidney/pathology , Male , Metabolic Clearance Rate , Mitochondria , Rats, Sprague-Dawley , Toxicokinetics
7.
J Biol Chem ; 294(8): 2949-2960, 2019 02 22.
Article in English | MEDLINE | ID: mdl-30635401

ABSTRACT

Protein disulfide bonds link pairs of cysteine residues in polypeptide chains. Many of these bonds serve a purely structural or energetic role, but a growing subset of cleavable disulfide bonds has been shown to control the function of the mature protein in which they reside. These allosteric disulfides and the factors that cleave these bonds are being identified across biological systems and life forms and have been shown to control hemostasis, the immune response, and viral infection in mammals. The discovery of these functional disulfides and a rationale for their facile nature has been aided by the emergence of a conformational signature for allosteric bonds. This post-translational modification mostly occurs extracellularly, making these chemical events prime drug targets. Indeed, a membrane-impermeable inhibitor of one of the cleaving factors is currently being trialed as an antithrombotic agent in cancer patients. Allosteric disulfides are firmly established as a sophisticated means by which a protein's shape and function can be altered; however, the full scope of this biological regulation will not be realized without new tools and techniques to study this regulation and innovative ways of targeting it.


Subject(s)
Hemostasis , Immunity , Neoplasm Proteins/immunology , Neoplasms/immunology , Protein Processing, Post-Translational/immunology , Virus Diseases/immunology , Animals , Humans , Neoplasms/pathology , Virus Diseases/pathology
8.
J Biol Chem ; 294(13): 4878-4888, 2019 03 29.
Article in English | MEDLINE | ID: mdl-30670593

ABSTRACT

ER protein 57 (ERp57), a thiol isomerase secreted from vascular cells, is essential for complete thrombus formation in vivo, but other extracellular ERp57 functions remain unexplored. Here, we employed a kinetic substrate-trapping approach to identify extracellular protein substrates of ERp57 in platelet-rich plasma. MS-based identification with immunochemical confirmation combined with gene ontology enrichment analysis revealed that ERp57 targets, among other substrates, components of the lectin pathway of complement activation: mannose-binding lectin, ficolin-2, ficolin-3, collectin-10, collectin-11, mannose-binding lectin-associated serine protease-1, and mannose-binding lectin-associated serine protease-2. Ficolin-3, the most abundant lectin pathway initiator in humans, circulates as disulfide-linked multimers of a monomer. ERp57 attenuated ficolin-3 ligand recognition and complement activation by cleaving intermolecular disulfide bonds in large ficolin-3 multimers, thereby reducing multimer size and ligand-binding affinity. We used MS to identify the disulfide-bonding pattern in ficolin-3 multimers and the disulfide bonds targeted by ERp57 and found that Cys6 and Cys23 in the N-terminal region of ficolin-3 form the intermolecular disulfide bonds in ficolin-3 multimers that are reduced by ERp57. Our results not only demonstrate that ERp57 can negatively regulate complement activation, but also identify a control mechanism for lectin pathway initiation in the vasculature. We conclude that extensive multimerization in large ficolin-3 multimers leads to a high affinity for ligands and strong complement-activating potential and that ERp57 suppresses complement activation by cleaving disulfide bonds in ficolin-3 and reducing its multimer size.


Subject(s)
Complement Pathway, Mannose-Binding Lectin , Glycoproteins/metabolism , Lectins/metabolism , Protein Disulfide-Isomerases/metabolism , Protein Multimerization , Proteolysis , Glycoproteins/genetics , Humans , Lectins/genetics , Protein Disulfide-Isomerases/genetics
9.
Circulation ; 139(10): 1300-1319, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30586735

ABSTRACT

BACKGROUND: Platelet-neutrophil interactions contribute to vascular occlusion and tissue damage in thromboinflammatory disease. Platelet glycoprotein Ibα (GPIbα), a key receptor for the cell-cell interaction, is believed to be constitutively active for ligand binding. Here, we established the role of platelet-derived protein disulfide isomerase (PDI) in reducing the allosteric disulfide bonds in GPIbα and enhancing the ligand-binding activity under thromboinflammatory conditions. METHODS: Bioinformatic analysis identified 2 potential allosteric disulfide bonds in GPIbα. Agglutination assays, flow cytometry, surface plasmon resonance analysis, a protein-protein docking model, proximity ligation assays, and mass spectrometry were used to demonstrate a direct interaction between PDI and GPIbα and to determine a role for PDI in regulating GPIbα function and platelet-neutrophil interactions. Also, real-time microscopy and animal disease models were used to study the pathophysiological role of PDI-GPIbα signaling under thromboinflammatory conditions. RESULTS: Deletion or inhibition of platelet PDI significantly reduced GPIbα-mediated platelet agglutination. Studies using PDI-null platelets and recombinant PDI or Anfibatide, a clinical-stage GPIbα inhibitor, revealed that the oxidoreductase activity of platelet surface-bound PDI was required for the ligand-binding function of GPIbα. PDI directly bound to the extracellular domain of GPIbα on the platelet surface and reduced the Cys4-Cys17 and Cys209-Cys248 disulfide bonds. Real-time microscopy with platelet-specific PDI conditional knockout and sickle cell disease mice demonstrated that PDI-regulated GPIbα function was essential for platelet-neutrophil interactions and vascular occlusion under thromboinflammatory conditions. Studies using a mouse model of ischemia/reperfusion-induced stroke indicated that PDI-GPIbα signaling played a crucial role in tissue damage. CONCLUSIONS: Our results demonstrate that PDI-facilitated cleavage of the allosteric disulfide bonds tightly regulates GPIbα function, promoting platelet-neutrophil interactions, vascular occlusion, and tissue damage under thromboinflammatory conditions.


Subject(s)
Anemia, Sickle Cell/enzymology , Blood Platelets/enzymology , Inflammation/enzymology , Neutrophils/metabolism , Platelet Adhesiveness , Platelet Glycoprotein GPIb-IX Complex/metabolism , Protein Disulfide-Isomerases/metabolism , Thrombosis/enzymology , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/genetics , Animals , Disease Models, Animal , Hemoglobins/genetics , Hemoglobins/metabolism , Humans , Inflammation/blood , Inflammation/genetics , Ligands , Mice, Inbred C57BL , Mice, Knockout , Platelet Glycoprotein GPIb-IX Complex/genetics , Protein Binding , Protein Disulfide-Isomerases/deficiency , Protein Disulfide-Isomerases/genetics , Signal Transduction , Thrombosis/blood , Thrombosis/genetics
10.
Molecules ; 25(23)2020 Dec 04.
Article in English | MEDLINE | ID: mdl-33291690

ABSTRACT

The polypeptide backbone of proteins is held together by two main types of covalent bonds: the peptide bonds that link the amino acid residues and the disulfide bonds that link pairs of cysteine amino acids. Disulfide bonds form as a protein folds in the cell and formation was assumed to be complete when the mature protein emerges. This is not the case for some secreted human blood proteins. The blood clotting protein, fibrinogen, and the protease inhibitor, α2-macroglobulin, exist in multiple disulfide-bonded or covalent states in the circulation. Thousands of different states are predicted assuming no dependencies on disulfide bond formation. In this study, probabilities for disulfide bond formation are employed to estimate numbers of covalent states of a model polypeptide with reference to α2-macroglobulin. When disulfide formation is interdependent in a protein, the number of covalent states is greatly reduced. Theoretical estimates of the number of states will aid the conceptual and experimental challenges of investigating multiple disulfide-bonded states of a protein.


Subject(s)
Blood Proteins/chemistry , Disulfides/chemistry , Cysteine/chemistry , Cystine/chemistry , Fibrinogen/chemistry , Globulins/chemistry , Humans , Peptides/chemistry , Probability , Protease Inhibitors/chemistry , Protein Conformation , Protein Folding
11.
J Biol Chem ; 293(46): 18010-18011, 2018 11 16.
Article in English | MEDLINE | ID: mdl-30446601

ABSTRACT

In the vertebrate eye, limiting oxidation of proteins and lipids is key to maintaining lens function and avoiding cataract formation. A study by Serebryany et al. identifies a surprising contributor to the eye's oxidative defense in their demonstration that γD-crystallin (HγD) functions as an oxidoreductase and uses disulfide exchange to initiate aggregation of mutant crystallins that mimic oxidative damage. These insights suggest a mechanism by which a dynamic pool of closely packed proteins might avoid oxidation-driven protein-folding traps, providing new avenues to understand the basis of a human disease with global impact.


Subject(s)
Disulfides/metabolism , Lens, Crystalline/metabolism , Oxidoreductases Acting on Sulfur Group Donors/metabolism , gamma-Crystallins/metabolism , Amino Acid Substitution , Cataract/physiopathology , Cysteine/chemistry , Humans , Mutation , Oxidation-Reduction , Oxidoreductases Acting on Sulfur Group Donors/genetics , gamma-Crystallins/genetics
12.
Bioconjug Chem ; 29(2): 546-558, 2018 02 21.
Article in English | MEDLINE | ID: mdl-29346731

ABSTRACT

PENAO (4-(N-(S-penicillaminylacetyl)amino) phenylarsonous acid), which is a mitochondria inhibitor that reacts with adenine nucleotide translocator (ANT), is currently being trialed in patients with solid tumors. To increase the stability of the drug, the formation of nanoparticles has been proposed. Herein, the direct synthesis of polymeric micelles based on the anticancer drug PENAO is presented. PENAO is readily available for amidation reaction to form PENAO MA (4-(N-(S-penicillaminylacetyl) amino) phenylarsonous acid methacrylamide) which undergoes RAFT (reversible addition-fragmentation chain transfer) polymerization with poly(ethylene glycol methyl ether methacrylate) as comonomer and poly(methyl methacrylate) (pMMA) as chain transfer agent, resulting in p(MMA)-b-p(PEG-co-PENAO) block copolymers with 3-15 wt % of PENAO MA. The different block copolymers self-assembled into micelle structures, varying in size and stability (Dh = 84-234 nm, cmc = 0.5-82 µg mol-1) depending on the hydrophilic to hydrophobic ratio of the polymer blocks and the amount of drug in the corona of the particle. The more stable micelle structures were investigated toward 143B human osteosarcoma cells, showing an enhanced cytotoxicity and cellular uptake compared to the free drug PENAO (IC50 (PENAO) = 2.7 ± 0.3 µM; IC50 (micelle M4) = 0.8 ± 0.02 µM). Furthermore, PENAOs arsonous acid residue remains active when incorporated into a polymer matrix and conjugates to small mono and closely spaced dithiols and is able to actively target the mitochondria, which is PENAO's main target to introduce growth inhibition in cancer cells. As a result, no cleavable linker between drug and polymer was necessary for the delivery of PENAO to osteosarcoma cells. These findings provide a rationale for in vivo studies of micelle M4 versus PENAO in an osteosarcoma animal model.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Arsenicals/chemistry , Arsenicals/pharmacology , Nanoparticles/chemistry , Polymers/chemistry , Polymers/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Carriers/chemistry , Humans , Mitochondria/drug effects , Mitochondria/pathology , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Polymerization , Sulfhydryl Compounds/chemistry
13.
Biophys J ; 112(1): 78-86, 2017 Jan 10.
Article in English | MEDLINE | ID: mdl-28076818

ABSTRACT

Tissue factor (TF) is a transmembrane glycoprotein that plays distinct roles in the initiation of extrinsic coagulation cascade and thrombosis. TF contains two disulfide bonds, one each in the N-terminal and C-terminal extracellular domains. The C-domain disulfide, Cys186-Cys209, has a -RHStaple configuration in crystal structures, suggesting that this disulfide carries high pre-stress. The redox state of this disulfide has been proposed to regulate TF encryption/decryption. Ablating the N-domain Cys49-Cys57 disulfide bond was found to increase the redox potential of the Cys186-Cys209 bond, implying an allosteric communication between the domains. Using molecular dynamics simulations, we observed that the Cys186-Cys209 disulfide bond retained the -RHStaple configuration, whereas the Cys49-Cys57 disulfide bond fluctuated widely. The Cys186-Cys209 bond featured the typical -RHStaple disulfide properties, such as a longer S-S bond length, larger C-S-S angles, and higher bonded prestress, in comparison to the Cys49-Cys57 bond. Force distribution analysis was used to sense the subtle structural changes upon ablating the disulfide bonds, and allowed us to identify a one-way allosteric communication mechanism from the N-terminal to the C-terminal domain. We propose a force propagation pathway using a shortest-pathway algorithm, which we suggest is a useful method for searching allosteric signal transduction pathways in proteins. As a possible explanation for the pathway being one-way, we identified a pronounced lower degree of conformational fluctuation, or effectively higher stiffness, in the N-terminal domain. Thus, the changes of the rigid domain (N-terminal domain) can induce mechanical force propagation to the soft domain (C-terminal domain), but not vice versa.


Subject(s)
Disulfides/chemistry , Molecular Dynamics Simulation , Thromboplastin/chemistry , Allosteric Regulation , Amino Acid Sequence , Biomechanical Phenomena , Mutation , Protein Domains , Thromboplastin/genetics , Thromboplastin/metabolism
14.
Blood ; 126(26): 2852-62, 2015 Dec 24.
Article in English | MEDLINE | ID: mdl-26474813

ABSTRACT

A subpopulation of platelets fulfills a procoagulant role in hemostasis and thrombosis by enabling the thrombin burst required for fibrin formation and clot stability at the site of vascular injury. Excess procoagulant activity is linked with pathological thrombosis. The identity of the procoagulant platelet has been elusive. The cell death marker 4-[N-(S-glutathionylacetyl)amino]phenylarsonous acid (GSAO) rapidly enters a subpopulation of agonist-stimulated platelets via an organic anion-transporting polypeptide and is retained in the cytosol through covalent reaction with protein dithiols. Labeling with GSAO, together with exposure of P-selectin, distinguishes necrotic from apoptotic platelets and correlates with procoagulant potential. GSAO(+) platelets form in occluding murine thrombi after ferric chloride injury and are attenuated with megakaryocyte-directed deletion of the cyclophilin D gene. These platelets form a procoagulant surface, supporting fibrin formation, and reduction in GSAO(+) platelets is associated with reduction in platelet thrombus size and fibrin formation. Analysis of platelets from human subjects receiving aspirin therapy indicates that these procoagulant platelets form despite aspirin therapy, but are attenuated by inhibition of the necrosis pathway. These findings indicate that the major subpopulation of platelets involved in fibrin formation are formed via regulated necrosis involving cyclophilin D, and that they may be targeted independent of platelet activation.


Subject(s)
Blood Platelets/metabolism , Platelet Activation/physiology , Thrombosis/metabolism , Animals , Arsenicals , Biomarkers/analysis , Cells, Cultured , Cyclophilins/metabolism , Flow Cytometry , Glutathione/analogs & derivatives , Humans , Mice , Microscopy, Confocal , Necrosis/metabolism
15.
Blood ; 133(3): 188-190, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30655304
16.
Blood ; 123(13): 2000-7, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24523239

ABSTRACT

Most proteins in nature are chemically modified after they are made to control how, when, and where they function. The 3 core features of proteins are posttranslationally modified: amino acid side chains can be modified, peptide bonds can be cleaved or isomerized, and disulfide bonds can be cleaved. Cleavage of peptide bonds is a major mechanism of protein control in the circulation, as exemplified by activation of the blood coagulation and complement zymogens. Cleavage of disulfide bonds is emerging as another important mechanism of protein control in the circulation. Recent advances in our understanding of control of soluble blood proteins and blood cell receptors by functional disulfide bonds is discussed as is how these bonds are being identified and studied.


Subject(s)
Allosteric Regulation/physiology , Blood Proteins/chemistry , Blood Proteins/metabolism , Disulfides/chemistry , Angiotensinogen/chemistry , Angiotensinogen/metabolism , Animals , Disulfides/metabolism , Humans , Hydrogen Bonding , Interleukin Receptor Common gamma Subunit/chemistry , Interleukin Receptor Common gamma Subunit/metabolism , Plasminogen/chemistry , Plasminogen/metabolism , beta 2-Glycoprotein I/chemistry , beta 2-Glycoprotein I/metabolism
17.
J Biol Chem ; 289(21): 15035-43, 2014 May 23.
Article in English | MEDLINE | ID: mdl-24700462

ABSTRACT

Protein translation is initiated with methionine in eukaryotes, and the majority of proteins have their N-terminal methionine removed by methionine aminopeptidases (MetAP1 and MetAP2) prior to action. Methionine removal can be important for protein function, localization, or stability. No mechanism of regulation of MetAP activity has been identified. MetAP2, but not MetAP1, contains a single Cys(228)-Cys(448) disulfide bond that has an -RHStaple configuration and links two ß-loop structures, which are hallmarks of allosteric disulfide bonds. From analysis of crystal structures and using mass spectrometry and activity assays, we found that the disulfide bond exists in oxidized and reduced states in the recombinant enzyme. The disulfide has a standard redox potential of -261 mV and is efficiently reduced by the protein reductant, thioredoxin, with a rate constant of 16,180 m(-1) s(-1). The MetAP2 disulfide bond also exists in oxidized and reduced states in glioblastoma tumor cells, and stressing the cells by oxygen or glucose deprivation results in more oxidized enzyme. The Cys(228)-Cys(448) disulfide is at the rim of the active site and is only three residues distant from the catalytic His(231), which suggested that cleavage of the bond would influence substrate hydrolysis. Indeed, oxidized and reduced isoforms have different catalytic efficiencies for hydrolysis of MetAP2 peptide substrates. These findings indicate that MetAP2 is post-translationally regulated by an allosteric disulfide bond, which controls substrate specificity and catalytic efficiency.


Subject(s)
Aminopeptidases/metabolism , Metalloendopeptidases/metabolism , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Aminopeptidases/classification , Aminopeptidases/genetics , Animals , Biocatalysis , Cell Line , Cell Line, Tumor , Crystallization , Disulfides/chemistry , Disulfides/metabolism , Electrophoresis, Polyacrylamide Gel , Glioblastoma/enzymology , Glioblastoma/pathology , Humans , Hydrolysis , Kinetics , Metalloendopeptidases/classification , Metalloendopeptidases/genetics , Models, Molecular , Oxidation-Reduction , Peptides/metabolism , Phylogeny , Substrate Specificity , Tandem Mass Spectrometry , Thioredoxins/metabolism
18.
J Biol Chem ; 289(5): 2992-3000, 2014 Jan 31.
Article in English | MEDLINE | ID: mdl-24338014

ABSTRACT

Plasma plasminogen is the precursor of the tumor angiogenesis inhibitor, angiostatin. Generation of angiostatin in blood involves activation of plasminogen to the serine protease plasmin and facilitated cleavage of two disulfide bonds and up to three peptide bonds in the kringle 5 domain of the protein. The mechanism of reduction of the two allosteric disulfides has been explored in this study. Using thiol-alkylating agents, mass spectrometry, and an assay for angiostatin formation, we show that the Cys(462)-Cys(541) disulfide bond is already cleaved in a fraction of plasma plasminogen and that this reduced plasminogen is the precursor for angiostatin formation. From the crystal structure of plasminogen, we propose that plasmin ligands such as phosphoglycerate kinase induce a conformational change in reduced kringle 5 that leads to attack by the Cys(541) thiolate anion on the Cys(536) sulfur atom of the Cys(512)-Cys(536) disulfide bond, resulting in reduction of the bond by thiol/disulfide exchange. Cleavage of the Cys(512)-Cys(536) allosteric disulfide allows further conformational change and exposure of the peptide backbone to proteolysis and angiostatin release. The Cys(462)-Cys(541) and Cys(512)-Cys(536) disulfides have -/+RHHook and -LHHook configurations, respectively, which are two of the 20 different measures of the geometry of a disulfide bond. Analysis of the structures of the known allosteric disulfide bonds identified six other bonds that have these configurations, and they share some functional similarities with the plasminogen disulfides. This suggests that the -/+RHHook and -LHHook disulfides, along with the -RHStaple bond, are potential allosteric configurations.


Subject(s)
Angiostatins/metabolism , Disulfides/metabolism , Fibrinolysin/metabolism , Plasminogen/metabolism , Protein Precursors/metabolism , Allosteric Regulation , Angiostatins/chemistry , Cysteine/chemistry , Cysteine/metabolism , Disulfides/chemistry , Fibrinolysin/chemistry , Humans , Oxidation-Reduction , Plasminogen/chemistry , Protein Precursors/chemistry , Protein Structure, Tertiary , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/metabolism
19.
J Am Chem Soc ; 137(12): 4215-22, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25794267

ABSTRACT

The entropy-driven affinity of trivalent (in)organic arsenicals for closely spaced dithiols has been exploited to develop a novel route to peptide/protein-polymer conjugation. A trivalent arsenous acid (As(III)) derivative (1) obtained from p-arsanilic acid (As(V)) was shown to readily undergo conjugation to the therapeutic peptide salmon calcitonin (sCT) via bridging of the Cys(1)-Cys(7) disulfide, which was verified by RP-HPLC and MALDI-ToF-MS. Conjugation was shown to proceed rapidly (t < 2 min) in situ and stoichiometrically through sequential reduction-conjugation protocols, therefore exhibiting conjugation efficiencies equivalent to those reported for the current leading disulfide-bond targeting strategies. Furthermore, using bovine serum albumin as a model protein, the trivalent organic arsenical 1 was found to demonstrate enhanced specificity for disulfide-bond bridging in the presence of free cysteine residues relative to established maleimide functional reagents. This specificity represents a shift toward potential orthogonality, by clearly distinguishing between the reactivity of mono- and disulfide-derived (vicinal or neighbors-through-space) dithiols. Finally, p-arsanilic acid was transformed into an initiator for aqueous single electron-transfer living radical polymerization, allowing the synthesis of hydrophilic arsenic-functional polymers which were shown to exhibit negligible cytotoxicity relative to a small molecule organic arsenical, and an unfunctionalized polymer control. Poly(poly[ethylene glycol] methyl ether acrylate) (PPEGA480, DPn = 10, Mn,NMR = 4900 g·mol(-1), D = 1.07) possessing a pentavalent arsenic acid (As(V)) α-chain end was transformed into trivalent As(III) post-polymerization via initial reduction by biological reducing agent glutathione (GSH), followed by binding of GSH. Conjugation of the resulting As(III)-functional polymer to sCT was realized within 35 min as indicated by RP-HPLC and verified later by thermodynamically driven release of sCT, from the conjugate, in the presence of strong chelating reagent ethanedithiol.


Subject(s)
Arsenicals/chemistry , Calcitonin/chemistry , Cysteine/chemistry , Acrylates/chemistry , Animals , Arsenicals/chemical synthesis , Arsenites/chemical synthesis , Arsenites/chemistry , Cell Line , Mice , Models, Molecular , Polyethylene Glycols/chemistry , Polymerization , Salmon , Sulfhydryl Compounds/chemistry
20.
Semin Thromb Hemost ; 41(7): 765-73, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26408919

ABSTRACT

Protein disulfide isomerase (PDI) is a 57-kDa oxidoreductase that facilitates cysteine thiol reactions inside and outside the cell. It mediates reduction or oxidation of protein disulfide bonds, thiol/disulfide exchange reactions, and transfer of NO from one protein thiol to another. It also has chaperone properties. PDI is actively secreted by most, if not all, of the cell types involved in thrombosis, binds to integrins on the cell surface, and circulates as a soluble protein in blood. It plays a critical role in thrombosis in mice and presumably the same role in human thrombosis. Eight proteins involved in thrombosis have been identified as PDI substrates; however, the role of this oxidoreductase in this process is not fully understood. Novel small-molecule PDI inhibitors have been developed and are being evaluated as antithrombotics in clinical trials. This combination of ongoing laboratory and clinical studies will greatly accelerate the pace of discovery and targeting of PDI function in thrombosis.


Subject(s)
Nitric Oxide/blood , Protein Disulfide-Isomerases/blood , Thrombosis/blood , Animals , Enzyme Inhibitors/therapeutic use , Humans , Mice , Oxidation-Reduction , Protein Disulfide-Isomerases/antagonists & inhibitors , Thrombosis/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL