1
|
Sivananthan S, Ahmed SA, Baig AM, Bhakta V, Sheffield WP. Substitution of reactive centre loop residues from C1 esterase inhibitor increases the inhibitory specificity of alpha-1 antitrypsin for plasma kallikrein. J Biotechnol 2025; 405:205-214. [PMID: 40441442 DOI: 10.1016/j.jbiotec.2025.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 04/21/2025] [Accepted: 05/18/2025] [Indexed: 06/02/2025]
Abstract
C1 esterase inhibitor (C1INH) is a member of the serpin superfamily of proteins and controls plasma kallikrein (Pka). Purified C1INH concentrates are effective in controlling C1INH deficiency (hereditary angioedema, HAE). Because C1INH is a relatively slow inhibitor of Pka, we sought to develop a more effective inhibitor by exchanging reactive centre loop (RCL) residues in another serpin, alpha-1 antitrypsin (AAT) variant M358R, with the corresponding residues of C1INH. Novel, soluble, N-terminally hexahistidine-tagged variants were expressed in E. coli, purified by nickel chelate chromatography, and characterized kinetically. AAT/C1INH loop exchange mutants were designated by the RCL residues exchanged using the reactive centre P1-P1' convention. Maximal exchange mutant AC (10-4') inhibited Pka 78-fold and activated Factor XI (FXIa) 350-fold less rapidly than AAT M358R. Eleven additional variants were expressed, restoring AAT residues stepwise. The most selective variant was AC (10-3/4'), which restored AAT residues from P2-P3' compared to AC (10-4'), and inhibited Pka 1.9-fold more rapidly, and FXIa 1.6-fold less rapidly, for a gain in selectivity of 2.8-fold (p < 0.0001), without increasing the stoichiometry of inhibition (SI). The most active variant was AC (10-3), in which both the rate of Pka and FXIa inhibition were elevated relative to AAT M358R values, without SI elevation. Other variants exhibited slower reaction rates and/or elevated SI values. These results indicate that RCL exchanges can be productively employed to change serpin specificity and selectivity, but that the most effective exchanges may not be contiguous due to cooperativity between RCL residues.
Collapse
Affiliation(s)
- Sangavi Sivananthan
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - S Ameer Ahmed
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Ammaar M Baig
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Varsha Bhakta
- Canadian Blood Services, Innovation and Portfolio Management, Medical Affairs and Innovation, Hamilton, Ontario, Canada
| | - William P Sheffield
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada; Canadian Blood Services, Innovation and Portfolio Management, Medical Affairs and Innovation, Hamilton, Ontario, Canada.
| |
Collapse
|
2
|
Atsavapranee B, Sunden F, Herschlag D, Fordyce P. Quantifying protein unfolding kinetics with a high-throughput microfluidic platform. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.15.633299. [PMID: 39868203 PMCID: PMC11761748 DOI: 10.1101/2025.01.15.633299] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Even after folding, proteins transiently sample unfolded or partially unfolded intermediates, and these species are often at risk of irreversible alteration (e.g. via proteolysis, aggregation, or post-translational modification). Kinetic stability, in addition to thermodynamic stability, can directly impact protein lifetime, abundance, and the formation of alternative, sometimes disruptive states. However, we have very few measurements of protein unfolding rates or how mutations alter these rates, largely due to technical challenges associated with their measurement. To address this need, we developed SPARKfold (Simultaneous Proteolysis Assay Revealing Kinetics of Folding), a microfluidic platform to express, purify, and measure unfolding rate constants for >1000 protein variants in parallel via on-chip native proteolysis. To demonstrate the power and potential of SPARKfold, we determined unfolding rate constants for 1,104 protein samples in parallel. We built a library of 31 dihydrofolate reductase (DHFR) orthologs with up to 78 chamber replicates per variant to provide the statistical power required to evaluate the system's ability to resolve subtle effects. SPARKfold rate constants for 5 constructs agreed with those obtained using traditional techniques across a 150-fold range, validating the accuracy of the technique. Comparisons of mutant kinetic effects via SPARKfold with previously published measurements impacts on folding thermodynamics provided information about the folding transition state and pathways via φ analysis. Overall, SPARKfold enables rapid characterization of protein variants to dissect the nature of the unfolding transition state. In future work, SPARKfold can reveal mutations that drive misfolding and aggregation and enable rational design of kinetically hyperstable variants for industrial use in harsh environments.
Collapse
Affiliation(s)
- B. Atsavapranee
- Department of Bioengineering, Stanford University, Stanford, CA 94305
| | - F. Sunden
- Department of Biochemistry, Stanford University, Stanford, CA 94305
| | - D. Herschlag
- Department of Biochemistry, Stanford University, Stanford, CA 94305
| | - P.M. Fordyce
- Department of Bioengineering, Stanford University, Stanford, CA 94305
- Department of Genetics, Stanford University, Stanford, CA 94305
- Sarafan ChEM-H Institute, Stanford University, Stanford, CA 94305
- Chan Zuckerberg Biohub, Stanford University, Stanford, CA 94305
| |
Collapse
|
3
|
Sivananthan S, Bhakta V, Chaechi Tehrani N, Sheffield WP. Prolonging the circulatory half-life of C1 esterase inhibitor via albumin fusion. PLoS One 2024; 19:e0305719. [PMID: 39441778 PMCID: PMC11498661 DOI: 10.1371/journal.pone.0305719] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 06/04/2024] [Indexed: 10/25/2024] Open
Abstract
Hereditary Angioedema (HAE) is an autosomal dominant disease characterized by episodic swelling, arising from genetic deficiency in C1-esterase inhibitor (C1INH), a regulator of several proteases including activated Plasma kallikrein (Pka). Many existing C1INH treatments exhibit short circulatory half-lives, precluding prophylactic use. Hexahistidine-tagged truncated C1INH (trC1INH lacking residues 1-97) with Mutated N-linked Glycosylation Sites N216Q/N231Q/N330Q (H6-trC1INH(MGS)), its murine serum albumin (MSA) fusion variant (H6-trC1INH(MGS)-MSA), and H6-MSA were expressed in Pichia pastoris and purified via nickel-chelate chromatography. Following intravenous injection in mice, the mean terminal half-life of H6-trC1INH(MGS)-MSA was significantly increased versus that of H6-trC1INH(MGS), by 3-fold, while remaining ~35% less than that of H6-MSA. The extended half-life was achieved with minimal, but significant, reduction in the mean second order rate constant of Pka inhibition of H6-trC1INH(MGS)-MSA by 33% relative to that of H6-trC1INH(MGS). Our results validate albumin fusion as a viable strategy for half-life extension of a natural inhibitor and suggest that H6-trC1INH(MGS)-MSA is worthy of investigation in a murine model of HAE.
Collapse
Affiliation(s)
- Sangavi Sivananthan
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Varsha Bhakta
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
- Canadian Blood Services, Innovation and Portfolio Management, Hamilton, Ontario, Canada
| | - Negin Chaechi Tehrani
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - William P. Sheffield
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
- Canadian Blood Services, Innovation and Portfolio Management, Hamilton, Ontario, Canada
| |
Collapse
|
4
|
Kell DB, Pretorius E. Proteomic Evidence for Amyloidogenic Cross-Seeding in Fibrinaloid Microclots. Int J Mol Sci 2024; 25:10809. [PMID: 39409138 PMCID: PMC11476703 DOI: 10.3390/ijms251910809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/20/2024] Open
Abstract
In classical amyloidoses, amyloid fibres form through the nucleation and accretion of protein monomers, with protofibrils and fibrils exhibiting a cross-β motif of parallel or antiparallel β-sheets oriented perpendicular to the fibre direction. These protofibrils and fibrils can intertwine to form mature amyloid fibres. Similar phenomena can occur in blood from individuals with circulating inflammatory molecules (and also some originating from viruses and bacteria). Such pathological clotting can result in an anomalous amyloid form termed fibrinaloid microclots. Previous proteomic analyses of these microclots have shown the presence of non-fibrin(ogen) proteins, suggesting a more complex mechanism than simple entrapment. We thus provide evidence against such a simple entrapment model, noting that clot pores are too large and centrifugation would have removed weakly bound proteins. Instead, we explore whether co-aggregation into amyloid fibres may involve axial (multiple proteins within the same fibril), lateral (single-protein fibrils contributing to a fibre), or both types of integration. Our analysis of proteomic data from fibrinaloid microclots in different diseases shows no significant quantitative overlap with the normal plasma proteome and no correlation between plasma protein abundance and their presence in fibrinaloid microclots. Notably, abundant plasma proteins like α-2-macroglobulin, fibronectin, and transthyretin are absent from microclots, while less abundant proteins such as adiponectin, periostin, and von Willebrand factor are well represented. Using bioinformatic tools, including AmyloGram and AnuPP, we found that proteins entrapped in fibrinaloid microclots exhibit high amyloidogenic tendencies, suggesting their integration as cross-β elements into amyloid structures. This integration likely contributes to the microclots' resistance to proteolysis. Our findings underscore the role of cross-seeding in fibrinaloid microclot formation and highlight the need for further investigation into their structural properties and implications in thrombotic and amyloid diseases. These insights provide a foundation for developing novel diagnostic and therapeutic strategies targeting amyloidogenic cross-seeding in blood clotting disorders.
Collapse
Affiliation(s)
- Douglas B. Kell
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St., Liverpool L69 7ZB, UK
- The Novo Nordisk Foundation Centre for Biosustainability, Building 220, Søltofts Plads 200, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch 7602, South Africa
| | - Etheresia Pretorius
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St., Liverpool L69 7ZB, UK
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch 7602, South Africa
| |
Collapse
|
5
|
Salang R, Phadungsil W, Geadkaew-Krenc A, Grams R. Investigation of a Serine Protease Inhibitor Active in the Infectious Stage of the Human Liver Fluke Opisthorchis viverrini. Pathogens 2024; 13:678. [PMID: 39204278 PMCID: PMC11356884 DOI: 10.3390/pathogens13080678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/07/2024] [Accepted: 08/07/2024] [Indexed: 09/03/2024] Open
Abstract
Serine protease inhibitors (serpins) participate in the regulation of inflammation, blood coagulation, and complement activation in humans. This research aimed to identify and characterize such inhibitors of the human liver fluke Opisthorchis viverrini. Parasite proteins that might contribute to the modulation of host physiology are of particular interest, especially as chronic opisthorchiasis increases the risk of developing biliary cancer. BLAST was used to find hypothetical serpins predicted from the parasite genome data. RNA extraction and reverse transcriptase PCR were used to isolate a serpin cDNA and to determine developmental transcript abundance. The evolutionary relation to other trematode serpins was revealed by phylogenetic analysis. Recombinant serpin was expressed in Escherichia coli and used to test the immunoreactivity of human opisthorchiasis sera and the inhibition of human serine proteases. A substantial serpin family with high sequence divergence among the members was found in the genus Opisthorchis. A serpin, different from previously analyzed trematode serpins, was cloned. The transcript was only detected in metacercariae and newly excysted juveniles. Human opisthorchiasis sera showed statistically significant reactivity to recombinant serpin. The serpin caused moderate inhibition of thrombin and low inhibition of kallikrein and chymotrypsin. This parasite serpin could be further evaluated as a diagnostic tool for early infection. Kallikrein and thrombin are involved in fibrinolysis; therefore, further research should explore the effects of the parasite serpin on this process.
Collapse
Affiliation(s)
| | | | | | - Rudi Grams
- Graduate Program in Biomedical Sciences, Faculty of Allied Health Sciences, Thammasat University, Khlong Luang, Pathum Thani 12120, Thailand; (R.S.); (W.P.); (A.G.-K.)
| |
Collapse
|
6
|
Bai X, Gao J, Guan X, Narum DE, Fornis LB, Griffith DE, Gao B, Sandhaus RA, Huang H, Chan ED. Analysis of alpha-1-antitrypsin (AAT)-regulated, glucocorticoid receptor-dependent genes in macrophages reveals a novel host defense function of AAT. Physiol Rep 2024; 12:e16124. [PMID: 39016119 PMCID: PMC11252833 DOI: 10.14814/phy2.16124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/14/2024] [Accepted: 06/14/2024] [Indexed: 07/18/2024] Open
Abstract
Alpha-1-antitrypsin (AAT) plays a homeostatic role in attenuating excessive inflammation and augmenting host defense against microbes. We demonstrated previously that AAT binds to the glucocorticoid receptor (GR) resulting in significant anti-inflammatory and antimycobacterial consequences in macrophages. Our current investigation aims to uncover AAT-regulated genes that rely on GR in macrophages. We incubated control THP-1 cells (THP-1control) and THP-1 cells knocked down for GR (THP-1GR-KD) with AAT, performed bulk RNA sequencing, and analyzed the findings. In THP-1control cells, AAT significantly upregulated 408 genes and downregulated 376 genes. Comparing THP-1control and THP-1GR-KD cells, 125 (30.6%) of the AAT-upregulated genes and 154 (41.0%) of the AAT-downregulated genes were significantly dependent on GR. Among the AAT-upregulated, GR-dependent genes, CSF-2 that encodes for granulocyte-monocyte colony-stimulating factor (GM-CSF), known to be host-protective against nontuberculous mycobacteria, was strongly upregulated by AAT and dependent on GR. We further quantified the mRNA and protein of several AAT-upregulated, GR-dependent genes in macrophages and the mRNA of several AAT-downregulated, GR-dependent genes. We also discussed the function(s) of selected AAT-regulated, GR-dependent gene products largely in the context of mycobacterial infections. In conclusion, AAT regulated several genes that are dependent on GR and play roles in host immunity against mycobacteria.
Collapse
Affiliation(s)
- Xiyuan Bai
- Department of MedicineRocky Mountain Regional Veterans Affairs Medical CenterAuroraColoradoUSA
- Department of Academic AffairsNational Jewish HealthDenverColoradoUSA
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Junfeng Gao
- Department of Immunology and Genomic MedicineNational Jewish HealthDenverColoradoUSA
| | - Xiaoyu Guan
- Department of Biostatistics and InformaticsUniversity of Colorado School of Public Health Anschutz Medical CampusAuroraColoradoUSA
| | - Drew E. Narum
- Department of Academic AffairsNational Jewish HealthDenverColoradoUSA
| | | | - David E. Griffith
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
- Department of MedicineNational Jewish HealthDenverColoradoUSA
| | - Bifeng Gao
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Robert A. Sandhaus
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
- Department of MedicineNational Jewish HealthDenverColoradoUSA
| | - Hua Huang
- Department of Immunology and Genomic MedicineNational Jewish HealthDenverColoradoUSA
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Edward D. Chan
- Department of MedicineRocky Mountain Regional Veterans Affairs Medical CenterAuroraColoradoUSA
- Department of Academic AffairsNational Jewish HealthDenverColoradoUSA
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| |
Collapse
|
7
|
Amer R, Koriat A. Aqueous humor perturbations in chronic smokers: a proteomic study. Sci Rep 2024; 14:11279. [PMID: 38760463 PMCID: PMC11101467 DOI: 10.1038/s41598-024-62039-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 05/13/2024] [Indexed: 05/19/2024] Open
Abstract
The detrimental effects of smoking are multisystemic and its effects on the eye health are significant. Smoking is a strong risk factor for age-related nuclear cataract, age-related macular degeneration, glaucoma, delayed corneal epithelial healing and increased risk of cystoid macular edema in patients with intermediate uveitis among others. We aimed to characterize the aqueous humor (AH) proteome in chronic smokers to gain insight into its perturbations and to identify potential biomarkers for smoking-associated ocular pathologies. Compared to the control group, chronic smokers displayed 67 (37 upregulated, 30 downregulated) differentially expressed proteins (DEPs). Analysis of DEPs from the biological point of view revealed that they were proteins involved in complement activation, lymphocyte mediated immunity, innate immune response, cellular oxidant detoxification, bicarbonate transport and platelet degranulation. From the molecular function point of view, DEPs were involved in oxygen binding, oxygen carrier activity, hemoglobin binding, peptidase/endopeptidase/cysteine-type endopeptidase inhibitory activity. Several of the upregulated proteins were acute phase reactant proteins such as clusterin, alpha-2-HS-glycoprotein, fibrinogen, alpha-1-antitrypsin, C4b-binding protein and serum amyloid A-2. Further research should confirm if these proteins might serve as biomarkers or therapeutic target for smoking-associated ocular diseases.
Collapse
Affiliation(s)
- Radgonde Amer
- Department of Ophthalmology, Hadassah Medical Center, Jerusalem, Israel.
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Adi Koriat
- Department of Ophthalmology, Hadassah Medical Center, Jerusalem, Israel
| |
Collapse
|
8
|
Rosenfeld MA, Yurina LV, Gavrilina ES, Vasilyeva AD. Post-Translational Oxidative Modifications of Hemostasis Proteins: Structure, Function, and Regulation. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:S14-S33. [PMID: 38621742 DOI: 10.1134/s0006297924140025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 04/17/2024]
Abstract
Reactive oxygen species (ROS) are constantly generated in a living organism. An imbalance between the amount of generated reactive species in the body and their destruction leads to the development of oxidative stress. Proteins are extremely vulnerable targets for ROS molecules, which can cause oxidative modifications of amino acid residues, thus altering structure and function of intra- and extracellular proteins. The current review considers the effect of oxidation on the structural rearrangements and functional activity of hemostasis proteins: coagulation system proteins such as fibrinogen, prothrombin/thrombin, factor VII/VIIa; anticoagulant proteins - thrombomodulin and protein C; proteins of the fibrinolytic system such as plasminogen, tissue plasminogen activator and plasminogen activator inhibitor-1. Structure and function of the proteins, oxidative modifications, and their detrimental consequences resulting from the induced oxidation or oxidative stress in vivo are described. Possible effects of oxidative modifications of proteins in vitro and in vivo leading to disruption of the coagulation and fibrinolysis processes are summarized and systematized, and the possibility of a compensatory mechanism in maintaining hemostasis under oxidative stress is analyzed.
Collapse
Affiliation(s)
- Mark A Rosenfeld
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, 119334, Russia.
| | - Lyubov V Yurina
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Elizaveta S Gavrilina
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Alexandra D Vasilyeva
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, 119334, Russia
| |
Collapse
|
9
|
Xu J, Ye W, Yang TT, Yan T, Cai H, Zhou A, Yang Y. DNA accelerates the protease inhibition of a bacterial serpin chloropin. Front Mol Biosci 2023; 10:1157186. [PMID: 37065444 PMCID: PMC10090351 DOI: 10.3389/fmolb.2023.1157186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/20/2023] [Indexed: 03/31/2023] Open
Abstract
Serine protease inhibitors (Serpins) are the most widely distributed protease inhibitors in nature and have been identified from all kingdoms of life. Eukaryotic serpins are most abundant with their activities often subject to modulation by cofactors; however, little is known about the regulation of prokaryotic serpins. To address this, here we prepared a recombinant bacteria serpin, termed chloropin, derived from green sulfur bacteria Chlorobium limicola and solved its crystal structure at 2.2 Å resolution. This showed a canonical inhibitory serpin conformation of native chloropin with a surface-exposed reactive loop and a large central beta-sheet. Enzyme activity analysis showed that chloropin could inhibit multiple proteases, such as thrombin and KLK7 with second order inhibition rate constants at 2.5×104 M−1s−1 and 4.5×104 M−1s−1 respectively, consistent with its P1 arginine residue. Heparin could accelerate the thrombin inhibition by ∼17-fold with a bell-shaped dose-dependent curve as seen with heparin-mediated thrombin inhibition by antithrombin. Interestingly, supercoiled DNA could accelerate the inhibition of thrombin by chloropin by 74-fold, while linear DNA accelerated the reaction by 142-fold through a heparin-like template mechanism. In contrast, DNA did not affect the inhibition of thrombin by antithrombin. These results indicate that DNA is likely a natural modulator of chloropin protecting the cell from endogenous or exogenous environmental proteases, and prokaryotic serpins have diverged during evolution to use different surface subsites for activity modulation.
Collapse
Affiliation(s)
- Jiawei Xu
- Department of Bioengineering, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong, China
| | - Wei Ye
- Department of Preventive Dentistry, The Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ting Ting Yang
- Department of Preventive Dentistry, The Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Teng Yan
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiyan Cai
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Haiyan Cai, ; Aiwu Zhou, ; Yufeng Yang,
| | - Aiwu Zhou
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Haiyan Cai, ; Aiwu Zhou, ; Yufeng Yang,
| | - Yufeng Yang
- Department of Bioengineering, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong, China
- *Correspondence: Haiyan Cai, ; Aiwu Zhou, ; Yufeng Yang,
| |
Collapse
|
10
|
Murshed M, Salim M, Boyd BJ. Existing and emerging mitigation strategies for the prevention of accidental overdose from oral pharmaceutical products. Eur J Pharm Biopharm 2022; 180:201-211. [DOI: 10.1016/j.ejpb.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/30/2022] [Accepted: 10/01/2022] [Indexed: 11/15/2022]
|
11
|
Fé LXSGM, Cipolatti EP, Pinto MCC, Branco S, Nogueira FCS, Ortiz GMD, Pinheiro ADS, Manoel EA. Enzymes in the time of COVID-19: An overview about the effects in the human body, enzyme market, and perspectives for new drugs. Med Res Rev 2022; 42:2126-2167. [PMID: 35762498 PMCID: PMC9350392 DOI: 10.1002/med.21919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 01/27/2022] [Accepted: 06/08/2022] [Indexed: 12/11/2022]
Abstract
The rising pandemic caused by a coronavirus, resulted in a scientific quest to discover some effective treatments against its etiologic agent, the severe acute respiratory syndrome‐coronavirus 2 (SARS‐CoV‐2). This research represented a significant scientific landmark and resulted in many medical advances. However, efforts to understand the viral mechanism of action and how the human body machinery is subverted during the infection are still ongoing. Herein, we contributed to this field with this compilation of the roles of both viral and human enzymes in the context of SARS‐CoV‐2 infection. In this sense, this overview reports that proteases are vital for the infection to take place: from SARS‐CoV‐2 perspective, the main protease (Mpro) and papain‐like protease (PLpro) are highlighted; from the human body, angiotensin‐converting enzyme‐2, transmembrane serine protease‐2, and cathepsins (CatB/L) are pointed out. In addition, the influence of the virus on other enzymes is reported as the JAK/STAT pathway and the levels of lipase, enzymes from the cholesterol metabolism pathway, amylase, aspartate aminotransferase, alanine aminotransferase, lactate dehydrogenase, and glyceraldehyde 3‐phosphate dehydrogenase are also be disturbed in SARS‐CoV‐2 infection. Finally, this paper discusses the importance of detailed enzymatic studies for future treatments against SARS‐CoV‐2, and how some issues related to the syndrome treatment can create opportunities in the biotechnological market of enzymes and the development of new drugs.
Collapse
Affiliation(s)
- Luana Xavier Soares Gomes Moura Fé
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Centro de Ciências da Saúde (CCS), Universidade Federal do Rio de Janeiro (UFRJ)-Cidade Universitária, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eliane Pereira Cipolatti
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Centro de Ciências da Saúde (CCS), Universidade Federal do Rio de Janeiro (UFRJ)-Cidade Universitária, Rio de Janeiro, Rio de Janeiro, Brazil.,Departamento de Engenharia Química, Instituto de Tecnologia, Universidade Federal Rural do Rio de Janeiro (UFRRJ), Seropédica, Rio de Janeiro, Brazil
| | - Martina Costa Cerqueira Pinto
- Departamento de Bioquímica, Instituto de Química, Centro de Tecnologia (CT), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil.,Chemical Engineering Program, Instituto Alberto Luiz Coimbra de Pós-graduação e Pesquisa de Engenharia (COPPE), Centro de Tecnologia (CT), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Suema Branco
- Biofísica Ambiental, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde (CCS), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fábio César Sousa Nogueira
- Departamento de Bioquímica, Instituto de Química, Centro de Tecnologia (CT), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gisela Maria Dellamora Ortiz
- Departamento de Fármacos e Medicamentos, Faculdade de Farmácia, Centro de Ciências da Saúde (CCS), Universidade Federal do Rio de Janeiro (UFRJ)-Cidade Universitária, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Anderson de Sá Pinheiro
- Departamento de Bioquímica, Instituto de Química, Centro de Tecnologia (CT), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Evelin Andrade Manoel
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Centro de Ciências da Saúde (CCS), Universidade Federal do Rio de Janeiro (UFRJ)-Cidade Universitária, Rio de Janeiro, Rio de Janeiro, Brazil.,Departamento de Bioquímica, Instituto de Química, Centro de Tecnologia (CT), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
12
|
A Review of Alpha-1 Antitrypsin Binding Partners for Immune Regulation and Potential Therapeutic Application. Int J Mol Sci 2022; 23:ijms23052441. [PMID: 35269582 PMCID: PMC8910375 DOI: 10.3390/ijms23052441] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 02/06/2023] Open
Abstract
Alpha-1 antitrypsin (AAT) is the canonical serine protease inhibitor of neutrophil-derived proteases and can modulate innate immune mechanisms through its anti-inflammatory activities mediated by a broad spectrum of protein, cytokine, and cell surface interactions. AAT contains a reactive methionine residue that is critical for its protease-specific binding capacity, whereby AAT entraps the protease on cleavage of its reactive centre loop, neutralises its activity by key changes in its tertiary structure, and permits removal of the AAT-protease complex from the circulation. Recently, however, the immunomodulatory role of AAT has come increasingly to the fore with several prominent studies focused on lipid or protein-protein interactions that are predominantly mediated through electrostatic, glycan, or hydrophobic potential binding sites. The aim of this review was to investigate the spectrum of AAT molecular interactions, with newer studies supporting a potential therapeutic paradigm for AAT augmentation therapy in disorders in which a chronic immune response is strongly linked.
Collapse
|
13
|
Abstract
Albumin is abundant in serum but is also excreted at mucosal surfaces and enters tissues when inflammation increases vascular permeability. Host-associated opportunistic pathogens encounter albumin during commensalism and when causing infections. Considering the ubiquitous presence of albumin, we investigated its role in the pathogenesis of infections with the model human fungal pathogen, Candida albicans. Albumin was introduced in various in vitro models that mimic different stages of systemic or mucosal candidiasis, where it reduced the ability of C. albicans to damage host cells. The amphipathic toxin candidalysin mediates necrotic host cell damage induced by C. albicans. Using cellular and biophysical assays, we determined that albumin functions by neutralizing candidalysin through hydrophobic interactions. We discovered that albumin, similarly, can neutralize a variety of fungal (α-amanitin), bacterial (streptolysin O and staurosporin), and insect (melittin) hydrophobic toxins. These data suggest albumin as a defense mechanism against toxins, which can play a role in the pathogenesis of microbial infections. IMPORTANCE Albumin is the most abundant serum protein in humans. During inflammation, serum albumin levels decrease drastically, and low albumin levels are associated with poor patient outcome. Thus, albumin may have specific functions during infection. Here, we describe the ability of albumin to neutralize hydrophobic microbial toxins. We show that albumin can protect against damage induced by the pathogenic yeast C. albicans by neutralizing its cytolytic toxin candidalysin. These findings suggest that albumin is a toxin-neutralizing protein that may play a role during infections with toxin-producing microorganisms.
Collapse
|
14
|
Rey F. Structure-function relations of the SARS-CoV-2 spike protein and impact of mutations in the variants of concern. C R Biol 2021; 344:77-110. [PMID: 34213849 DOI: 10.5802/crbiol.53] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
This review covers the main features of the severe acquired respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein, its interaction with the main entry receptor, the human angiotensin converting enzyme 2 (ACE2), and the subsequent membrane fusion step. The focus is on the structural organization of these proteins and mechanistic aspects of their interactions that lead to cytoplasmic release of the viral genome. The most potently neutralizing antibodies against SARS-CoV-2 were shown to interfere with the spike/ACE2 interaction. I thus also review the location and the potential impact of mutations in the spike protein observed in the variants of concern that emerged concomitantly with acquired immunity in the population after one year of virus circulation. Understanding how these interactions affect the spike/ACE2 interactions and the subsequent spike-protein-induced membrane fusion reaction is important to stay one step ahead of the virus evolution and develop efficient countermeasures.
Collapse
Affiliation(s)
- Félix Rey
- Unité de Virologie Structurale, CNRS UMR 3569, Institut Pasteur, 28 rue du Dr Roux, 75015 Paris, France
| |
Collapse
|
15
|
Lim J, Lee K, Im H. Reinforcement of the Unfolded Protein Response Mitigates Cytotoxicity Induced by Human Z‐Type α
1
‐Antitrypsin. B KOREAN CHEM SOC 2021. [DOI: 10.1002/bkcs.12289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Jaeyeon Lim
- Department of Integrative Bioscience and Biotechnology Sejong University Seoul 05006 South Korea
| | - Kyunghee Lee
- Department of Chemistry Sejong University Seoul 05006 South Korea
| | - Hana Im
- Department of Integrative Bioscience and Biotechnology Sejong University Seoul 05006 South Korea
| |
Collapse
|
16
|
Identification of an alpha-1 antitrypsin variant with enhanced specificity for factor XIa by phage display, bacterial expression, and combinatorial mutagenesis. Sci Rep 2021; 11:5565. [PMID: 33692375 PMCID: PMC7946950 DOI: 10.1038/s41598-021-84618-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 02/15/2021] [Indexed: 01/20/2023] Open
Abstract
Coagulation Factor XIa (FXIa) is an emerging target for antithrombotic agent development. The M358R variant of the serpin alpha-1 antitrypsin (AAT) inhibits both FXIa and other proteases. Our aim was to enhance the specificity of AAT M358R for FXIa. We randomized two AAT M358R phage display libraries at reactive centre loop positions P13-P8 and P7-P3 and biopanned them with FXIa. A bacterial expression library randomized at P2′-P3′ was also probed. Resulting novel variants were expressed as recombinant proteins in E. coli and their kinetics of FXIa inhibition determined. The most potent FXIa-inhibitory motifs were: P13-P8, HASTGQ; P7-P3, CLEVE; and P2-P3′, PRSTE (respectively, novel residues bolded). Selectivity for FXIa over thrombin was increased up to 34-fold versus AAT M358R for these single motif variants. Combining CLEVE and PRSTE motifs in AAT-RC increased FXIa selectivity for thrombin, factors XIIa, Xa, activated protein C, and kallikrein by 279-, 143-, 63-, 58-, and 36-fold, respectively, versus AAT M358R. AAT-RC lengthened human plasma clotting times less than AAT M358R. AAT-RC rapidly and selectively inhibits FXIa and is worthy of testing in vivo. AAT specificity can be focused on one target protease by selection in phage and bacterial systems coupled with combinatorial mutagenesis.
Collapse
|
17
|
Gaddh M, Rosovsky RP. Venous Thromboembolism: Genetics and Thrombophilias. Semin Respir Crit Care Med 2021; 42:271-283. [PMID: 33694139 DOI: 10.1055/s-0041-1723937] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Venous thromboembolism (VTE) is a major cause of morbidity and mortality throughout the world. Up to one half of patients who present with VTE will have an underlying thrombophilic defect. This knowledge has led to a widespread practice of testing for such defects in patients who develop VTE. However, identifying a hereditary thrombophilia by itself does not necessarily change outcomes or dictate therapy. Furthermore, family history of VTE by itself can increase an asymptomatic person's VTE risk several-fold, independent of detecting a known inherited thrombophilia. In this article, we will describe the current validated hereditary thrombophilias including their history, prevalence, and association with VTE. With a focus on evaluating both risks and benefits of testing, we will also explore the controversies of why, who, and when to test as well as discuss contemporary societal guidelines. Lastly, we will share how these tests have been integrated into clinical practice and how to best utilize them in the future.
Collapse
Affiliation(s)
- Manila Gaddh
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia
| | - Rachel P Rosovsky
- Division of Hematology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
18
|
Kouroumalis E, Voumvouraki A, Augoustaki A, Samonakis DN. Autophagy in liver diseases. World J Hepatol 2021; 13:6-65. [PMID: 33584986 PMCID: PMC7856864 DOI: 10.4254/wjh.v13.i1.6] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 12/10/2020] [Accepted: 12/26/2020] [Indexed: 02/06/2023] Open
Abstract
Autophagy is the liver cell energy recycling system regulating a variety of homeostatic mechanisms. Damaged organelles, lipids and proteins are degraded in the lysosomes and their elements are re-used by the cell. Investigations on autophagy have led to the award of two Nobel Prizes and a health of important reports. In this review we describe the fundamental functions of autophagy in the liver including new data on the regulation of autophagy. Moreover we emphasize the fact that autophagy acts like a two edge sword in many occasions with the most prominent paradigm being its involvement in the initiation and progress of hepatocellular carcinoma. We also focused to the implication of autophagy and its specialized forms of lipophagy and mitophagy in the pathogenesis of various liver diseases. We analyzed autophagy not only in well studied diseases, like alcoholic and nonalcoholic fatty liver and liver fibrosis but also in viral hepatitis, biliary diseases, autoimmune hepatitis and rare diseases including inherited metabolic diseases and also acetaminophene hepatotoxicity. We also stressed the different consequences that activation or impairment of autophagy may have in hepatocytes as opposed to Kupffer cells, sinusoidal endothelial cells or hepatic stellate cells. Finally, we analyzed the limited clinical data compared to the extensive experimental evidence and the possible future therapeutic interventions based on autophagy manipulation.
Collapse
Affiliation(s)
- Elias Kouroumalis
- Liver Research Laboratory, University of Crete Medical School, Heraklion 71110, Greece
| | - Argryro Voumvouraki
- 1 Department of Internal Medicine, AHEPA University Hospital, Thessaloniki 54636, Greece
| | - Aikaterini Augoustaki
- Department of Gastroenterology and Hepatology, University Hospital of Crete, Heraklion 71110, Greece
| | - Dimitrios N Samonakis
- Department of Gastroenterology and Hepatology, University Hospital of Crete, Heraklion 71110, Greece.
| |
Collapse
|
19
|
Zhang X, Gao B, Xu B. Association between plasminogen activator inhibitor-1 (PAI-1) 4G/5G polymorphism and risk of Alzheimer's disease, metabolic syndrome, and female infertility: A meta-analysis. Medicine (Baltimore) 2020; 99:e23660. [PMID: 33327353 PMCID: PMC7738113 DOI: 10.1097/md.0000000000023660] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Plasminogen activator inhibitor-1 (PAI-1) is considered to be involved in the physiopathological mechanisms of Alzheimer's disease (AD), metabolic syndrome (MetS), and female infertility. Previous studies investigating the association between PAI-14G/5G (rs1799889) gene polymorphism and the risk of AD, MetS, and female infertility have reported inconsistent results. The aim of the present study was to investigate possible associations. METHODS Eligible studies were retrieved through PubMed, Medline, EMBASE, CNKI, and WANFANG databases. The odds ratios (ORs) and 95% confidence intervals (CIs) were used to assess the associations. Subgroup analyses by ethnicity and mean age, sensitivity analyses, and publication bias were performed. RESULTS Five studies (four articles) for AD, six studies (six articles) for MetS, and four studies (four articles) for female infertility were included in this meta-analysis. Our results showed no significant associations between the PAI-14G/5G polymorphism and the risk of AD and female infertility in five genetic models. For the risk of MetS, the PAI-1 4G/5G (rs1799889) polymorphism may be associated with the risk of MetS (4G vs 5G, OR = 1.31, 95%CI = 1.04-1.64, P = .021), especially in Asians (4G/4G vs 4G/5G+5G/5G, OR = 1.38, 95%CI = 1.01-1.87, P = .041) and patients with mean age > 50 years old (4G/4G vs 4G/5G+5G/5G, OR = 1.36, 95%CI = 1.03-1.78, P = .029). CONCLUSION The present meta-analysis suggested that the PAI-1 4G/5G polymorphism might be associated with the risk of MetS, but no evidence was detected for AD and female infertility.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Neurology, Shenyang First People's Hospital, Dadong District
| | - Bai Gao
- Department of Nerve Function, ShengJing Hospital of China Medical University, Heping District, Shenyang, Liaoning Province, People's Republic of China
| | - Bing Xu
- Department of Neurology, Shenyang First People's Hospital, Dadong District
| |
Collapse
|
20
|
Rezaie AR, Giri H. Anticoagulant and signaling functions of antithrombin. J Thromb Haemost 2020; 18:3142-3153. [PMID: 32780936 PMCID: PMC7855051 DOI: 10.1111/jth.15052] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/25/2020] [Accepted: 08/04/2020] [Indexed: 12/19/2022]
Abstract
Antithrombin (AT) is a major plasma glycoprotein of the serpin superfamily that regulates the proteolytic activity of the procoagulant proteases of both intrinsic and extrinsic pathways. Two important structural features that participate in the regulatory function of AT include a mobile reactive center loop that binds to active site of coagulation proteases, trapping them in the form of inactive covalent complexes, and a basic D-helix that binds to therapeutic heparins and heparan sulfate proteoglycans (HSPGs) on vascular endothelial cells. The binding of D-helix of AT by therapeutic heparins promotes the reactivity of the serpin with coagulation proteases by several orders of magnitude by both a conformational activation of the serpin and a template (bridging) mechanism. In addition to its essential anticoagulant function, AT elicits a potent anti-inflammatory signaling response when it binds to distinct vascular endothelial cell HSPGs, thereby inducing prostacyclin synthesis. Syndecans-4 has been found as a specific membrane-bound HSPG receptor on endothelial cells that relays the signaling effect of AT to the relevant second messenger molecules in the signal transduction pathways inside the cell. However, following cleavage by coagulation proteases and/or by spontaneous conversion to a latent form, AT loses both its anti-inflammatory activity and high-affinity interaction with heparin and HSPGs. Interestingly, these low-affinity heparin conformers of AT elicit potent proapoptotic and antiangiogenic activities by also binding to specific HSPGs by unknown mechanisms. This review article will summarize current knowledge about mechanisms through which different conformers of AT exert their serine protease inhibitory and intracellular signaling functions in these biological pathways.
Collapse
Affiliation(s)
- Alireza R. Rezaie
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Hemant Giri
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104
| |
Collapse
|
21
|
Factor XII/XIIa inhibitors: Their discovery, development, and potential indications. Eur J Med Chem 2020; 208:112753. [DOI: 10.1016/j.ejmech.2020.112753] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/10/2020] [Accepted: 08/10/2020] [Indexed: 12/21/2022]
|
22
|
The cytoplasmic tail of human mannosidase Man1b1 contributes to catalysis-independent quality control of misfolded alpha1-antitrypsin. Proc Natl Acad Sci U S A 2020; 117:24825-24836. [PMID: 32958677 DOI: 10.1073/pnas.1919013117] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The failure of polypeptides to achieve conformational maturation following biosynthesis can result in the formation of protein aggregates capable of disrupting essential cellular functions. In the secretory pathway, misfolded asparagine (N)-linked glycoproteins are selectively sorted for endoplasmic reticulum-associated degradation (ERAD) in response to the catalytic removal of terminal alpha-linked mannose units. Remarkably, ER mannosidase I/Man1b1, the first alpha-mannosidase implicated in this conventional N-glycan-mediated process, can also contribute to ERAD in an unconventional, catalysis-independent manner. To interrogate this functional dichotomy, the intracellular fates of two naturally occurring misfolded N-glycosylated variants of human alpha1-antitrypsin (AAT), Null Hong Kong (NHK), and Z (ATZ), in Man1b1 knockout HEK293T cells were monitored in response to mutated or truncated forms of transfected Man1b1. As expected, the conventional catalytic system requires an intact active site in the Man1b1 luminal domain. In contrast, the unconventional system is under the control of an evolutionarily extended N-terminal cytoplasmic tail. Also, N-glycans attached to misfolded AAT are not required for accelerated degradation mediated by the unconventional system, further demonstrating its catalysis-independent nature. We also established that both systems accelerate the proteasomal degradation of NHK in metabolic pulse-chase labeling studies. Taken together, these results have identified the previously unrecognized regulatory capacity of the Man1b1 cytoplasmic tail and provided insight into the functional dichotomy of Man1b1 as a component in the mammalian proteostasis network.
Collapse
|
23
|
Lüling R, Schmeißer W, Siegert M, Mückter H, Dietrich A, Thiermann H, Gudermann T, John H, Steinritz D. Identification of creatine kinase and alpha-1 antitrypsin as protein targets of alkylation by sulfur mustard. Drug Test Anal 2020; 13:268-282. [PMID: 32852113 DOI: 10.1002/dta.2916] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/16/2020] [Accepted: 08/18/2020] [Indexed: 12/18/2022]
Abstract
Sulfur mustard (SM) is a toxic chemical warfare agent deployed in several conflicts within the last 100 years and still represents a threat in terroristic attacks and warfare. SM research focuses on understanding the pathophysiology of SM and identifying novel biomarkers of exposure. SM is known to alkylate nucleophilic moieties of endogenous proteins, for example, free thiol groups of cysteine residues. The two-dimensional-thiol-differences in gel electrophoresis (2D-thiol-DIGE) technique is an initial proteomics approach to detect proteins with free cysteine residues. These amino acids are selectively labeled with infrared-maleimide dyes visualized after GE. Cysteine residues derivatized by alkylating agents are no longer accessible for the maleimide-thiol coupling resulting in the loss of the fluorescent signal of the corresponding protein. To prove the applicability of 2D-thiol-DIGE, this technology was exemplarily applied to neat human serum albumin treated with SM, to lysates from human cell culture exposed to SM as well as to human plasma exposed to CEES (chloroethyl ethyl sulfide, an SM analogue). Exemplarily, the most prominent proteins modified by SM were identified by matrix-assisted laser desorption/ionization time-of-flight (tandem) mass spectrometry, MALDI-TOF MS(/MS), as creatine kinase (CK) from human cells and as alpha-1 antitrypsin (A1AT) from plasma samples. Peptides containing the residue Cys282 of CK and Cys232 of A1AT were unambiguously identified by micro liquid chromatography-electrospray ionization high-resolution tandem-mass spectrometry (μLC-ESI MS/HR MS) as being alkylated by SM bearing the specific hydroxyethylthioethyl-(HETE)-moiety. Both peptides might represent potential biomarkers of SM exposure. This is the first report introducing these endogenous proteins as targets of SM alkylation.
Collapse
Affiliation(s)
- Robin Lüling
- Bundeswehr Institute of Pharmacology and Toxicology, Munich, Germany.,Walther-Straub-Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | | | - Markus Siegert
- Bundeswehr Institute of Pharmacology and Toxicology, Munich, Germany.,Department of Chemistry, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Harald Mückter
- Walther-Straub-Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Alexander Dietrich
- Walther-Straub-Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Horst Thiermann
- Bundeswehr Institute of Pharmacology and Toxicology, Munich, Germany
| | - Thomas Gudermann
- Walther-Straub-Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Harald John
- Bundeswehr Institute of Pharmacology and Toxicology, Munich, Germany
| | - Dirk Steinritz
- Bundeswehr Institute of Pharmacology and Toxicology, Munich, Germany.,Walther-Straub-Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität Munich, Munich, Germany.,Bundeswehr Medical Service Academy, Munich, Germany
| |
Collapse
|
24
|
Ma L, Wu J, Zheng Y, Shu Z, Wei Z, Sun Y, Carrell RW, Zhou A. Heparin Blocks the Inhibition of Tissue Kallikrein 1 by Kallistatin through Electrostatic Repulsion. Biomolecules 2020; 10:E828. [PMID: 32481593 PMCID: PMC7356578 DOI: 10.3390/biom10060828] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 05/26/2020] [Accepted: 05/26/2020] [Indexed: 11/16/2022] Open
Abstract
Kallistatin, also known as SERPINA4, has been implicated in the regulation of blood pressure and angiogenesis, due to its specific inhibition of tissue kallikrein 1 (KLK1) and/or by its heparin binding ability. The binding of heparin on kallistatin has been shown to block the inhibition of KLK1 by kallistatin but the detailed molecular mechanism underlying this blockade is unclear. Here we solved the crystal structures of human kallistatin and its complex with heparin at 1.9 and 1.8 Å resolution, respectively. The structures show that kallistatin has a conserved serpin fold and undergoes typical stressed-to-relaxed conformational changes upon reactive loop cleavage. Structural analysis and mutagenesis studies show that the heparin binding site of kallistatin is located on a surface with positive electrostatic potential near a unique protruded 310 helix between helix H and strand 2 of β-sheet C. Heparin binding on this site would prevent KLK1 from docking onto kallistatin due to the electrostatic repulsion between heparin and the negatively charged surface of KLK1, thus blocking the inhibition of KLK1 by kallistatin. Replacement of the acidic exosite 1 residues of KLK1 with basic amino acids as in thrombin resulted in accelerated inhibition. Taken together, these data indicate that heparin controls the specificity of kallistatin, such that kinin generation by KLK1 within the microcirculation will be locally protected by the binding of kallistatin to the heparin-like glycosaminoglycans of the endothelium.
Collapse
Affiliation(s)
- Lina Ma
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (L.M.); (J.W.); (Z.S.); (Z.W.)
| | - Jiawei Wu
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (L.M.); (J.W.); (Z.S.); (Z.W.)
| | - Ying Zheng
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (L.M.); (J.W.); (Z.S.); (Z.W.)
| | - Zimei Shu
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (L.M.); (J.W.); (Z.S.); (Z.W.)
| | - Zhenquan Wei
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (L.M.); (J.W.); (Z.S.); (Z.W.)
| | - Yinbiao Sun
- Randall Division of Cell & Molecular Biophysics, Faculty of Life Sciences & Medicine, King’s College London, New Hunt’s House, Guy’s Campus, London SE1 1UL, UK;
| | - Robin W. Carrell
- Department of Haematology, University of Cambridge, Cambridge CB2 0XY, UK;
| | - Aiwu Zhou
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (L.M.); (J.W.); (Z.S.); (Z.W.)
| |
Collapse
|
25
|
Juarez-Navarro K, Ayala-Garcia VM, Ruiz-Baca E, Meneses-Morales I, Rios-Banuelos JL, Lopez-Rodriguez A. Assistance for Folding of Disease-Causing Plasma Membrane Proteins. Biomolecules 2020; 10:biom10050728. [PMID: 32392767 PMCID: PMC7277483 DOI: 10.3390/biom10050728] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 04/16/2020] [Accepted: 04/21/2020] [Indexed: 02/06/2023] Open
Abstract
An extensive catalog of plasma membrane (PM) protein mutations related to phenotypic diseases is associated with incorrect protein folding and/or localization. These impairments, in addition to dysfunction, frequently promote protein aggregation, which can be detrimental to cells. Here, we review PM protein processing, from protein synthesis in the endoplasmic reticulum to delivery to the PM, stressing the main repercussions of processing failures and their physiological consequences in pathologies, and we summarize the recent proposed therapeutic strategies to rescue misassembled proteins through different types of chaperones and/or small molecule drugs that safeguard protein quality control and regulate proteostasis.
Collapse
|
26
|
Ke PY. Mitophagy in the Pathogenesis of Liver Diseases. Cells 2020; 9:cells9040831. [PMID: 32235615 PMCID: PMC7226805 DOI: 10.3390/cells9040831] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/25/2020] [Accepted: 03/27/2020] [Indexed: 02/07/2023] Open
Abstract
Autophagy is a catabolic process involving vacuolar sequestration of intracellular components and their targeting to lysosomes for degradation, thus supporting nutrient recycling and energy regeneration. Accumulating evidence indicates that in addition to being a bulk, nonselective degradation mechanism, autophagy may selectively eliminate damaged mitochondria to promote mitochondrial turnover, a process termed “mitophagy”. Mitophagy sequesters dysfunctional mitochondria via ubiquitination and cargo receptor recognition and has emerged as an important event in the regulation of liver physiology. Recent studies have shown that mitophagy may participate in the pathogenesis of various liver diseases, such as liver injury, liver steatosis/fatty liver disease, hepatocellular carcinoma, viral hepatitis, and hepatic fibrosis. This review summarizes the current knowledge on the molecular regulations and functions of mitophagy in liver physiology and the roles of mitophagy in the development of liver-related diseases. Furthermore, the therapeutic implications of targeting hepatic mitophagy to design a new strategy to cure liver diseases are discussed.
Collapse
Affiliation(s)
- Po-Yuan Ke
- Department of Biochemistry & Molecular Biology and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; ; Tel.: +886-3-211-8800 (ext. 5115); Fax: +886-3-211-8700
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- Division of Allergy, Immunology, and Rheumatology, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| |
Collapse
|
27
|
Lalonde ME, Koyuturk I, Brochu D, Jabbour J, Gilbert M, Durocher Y. Production of α2,6-sialylated and non-fucosylated recombinant alpha-1-antitrypsin in CHO cells. J Biotechnol 2020; 307:87-97. [PMID: 31697975 DOI: 10.1016/j.jbiotec.2019.10.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 10/23/2019] [Accepted: 10/29/2019] [Indexed: 01/05/2023]
Abstract
Alpha-1-antitrypsin (A1AT) is an abundant serum inhibitor of serine proteases. A1AT deficiency is a common genetic disorder which is currently treated with augmentation therapies. These treatments involve weekly injections of patients with purified plasma-derived A1AT. Such therapies can be extremely expensive and rely on plasma donors. Hence, large-scale production of recombinant A1AT (rA1AT) could greatly benefit these patients, as it could decrease the cost of treatments, reduce biosafety concerns and ensure quantitative and qualitative controls of the protein. In this report, we sought to produce α2,6-sialylated rA1AT with our cumate-inducible stable CHO pool expression system. Our different CHO pools could reach volumetric productivities of 1,2 g/L. The human α2,6-sialyltransferase was stably expressed in these cells in order to mimic elevated α2,6-sialylation levels of native A1AT protein. Sialylation of the recombinant protein was stable over the duration of the fed-batch production phase and was higher in a pool where cells were sorted and enriched by FACS based on cell-surface α2,6-sialylation. Addition of ManNAc to the cell culture media during production enhanced both α2,3 and α2,6 A1AT sialylation levels whereas addition of 2F-peracetylfucose potently inhibited fucosylation of the protein. Finally, we demonstrated that rA1AT proteins exhibited human neutrophil elastase inhibitory activities similar to the commercial human plasma-derived A1AT.
Collapse
Affiliation(s)
- Marie-Eve Lalonde
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Québec, H3C 3J7, Canada
| | - Izel Koyuturk
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Québec, H3C 3J7, Canada
| | - Denis Brochu
- Life Sciences, Human Health Therapeutics Research Centre, 100 Sussex Drive, National Research Council Canada, Ottawa, Ontario, K1A OR6, Canada
| | - Jonathan Jabbour
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Québec, H3C 3J7, Canada
| | - Michel Gilbert
- Life Sciences, Human Health Therapeutics Research Centre, 100 Sussex Drive, National Research Council Canada, Ottawa, Ontario, K1A OR6, Canada
| | - Yves Durocher
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Québec, H3C 3J7, Canada; Life Sciences, Human Health Therapeutics Research Centre, Building Montreal-Royalmount, National Research Council Canada, Montréal, Québec, H4P 2R2, Canada.
| |
Collapse
|
28
|
McElvaney OF, Murphy MP, Reeves EP, McElvaney NG. Anti-cytokines as a Strategy in Alpha-1 Antitrypsin Deficiency. CHRONIC OBSTRUCTIVE PULMONARY DISEASES-JOURNAL OF THE COPD FOUNDATION 2020; 7:203-213. [PMID: 32503090 DOI: 10.15326/jcopdf.7.3.2019.0171] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
For many years, the lung disease associated with alpha-1 antitrypsin (AAT) deficiency (AATD) was perceived as being secondary to an imbalance between this serine protease inhibitor and the target protease, neutrophil elastase (NE). More recently, a greater understanding of the pathways leading to lung inflammation has shed light on new potential attributes and presented AATD as an inflammatory condition in which proteases and neutrophils still play a major role, but in which pro-inflammatory cytokines, either induced by the actions of NE or by other pro-inflammatory processes normally modulated by AAT, are involved. In this review, we will look at the various cytokines centrally involved in AATD lung disease, and how a greater understanding of their contribution may help development of targeted therapies.
Collapse
Affiliation(s)
- Oisín F McElvaney
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - Mark P Murphy
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - Emer P Reeves
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - Noel G McElvaney
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
29
|
Scott BM, Sheffield WP. Engineering the serpin α 1 -antitrypsin: A diversity of goals and techniques. Protein Sci 2019; 29:856-871. [PMID: 31774589 DOI: 10.1002/pro.3794] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/19/2019] [Accepted: 11/19/2019] [Indexed: 12/19/2022]
Abstract
α1 -Antitrypsin (α1 -AT) serves as an archetypal example for the serine proteinase inhibitor (serpin) protein family and has been used as a scaffold for protein engineering for >35 years. Techniques used to engineer α1 -AT include targeted mutagenesis, protein fusions, phage display, glycoengineering, and consensus protein design. The goals of engineering have also been diverse, ranging from understanding serpin structure-function relationships, to the design of more potent or more specific proteinase inhibitors with potential therapeutic relevance. Here we summarize the history of these protein engineering efforts, describing the techniques applied to engineer α1 -AT, specific mutants of interest, and providing an appended catalog of the >200 α1 -AT mutants published to date.
Collapse
Affiliation(s)
- Benjamin M Scott
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland.,Biosystems and Biomaterials Division, National Institute of Standards and Technology, Gaithersburg, Maryland
| | - William P Sheffield
- Canadian Blood Services, Centre for Innovation, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
30
|
Affiliation(s)
- Y. Mine
- Department of Food Science, University of Guelph, Guelph, Ontario, Canada N1G 2W1
| | - P. Rupa
- Department of Food Science, University of Guelph, Guelph, Ontario, Canada N1G 2W1
| |
Collapse
|
31
|
Roudnický P, Vorel J, Ilgová J, Benovics M, Norek A, Jedličková L, Mikeš L, Potěšil D, Zdráhal Z, Dvořák J, Gelnar M, Kašný M. Identification and partial characterization of a novel serpin from Eudiplozoon nipponicum (Monogenea, Polyopisthocotylea). ACTA ACUST UNITED AC 2018; 25:61. [PMID: 30516130 PMCID: PMC6280883 DOI: 10.1051/parasite/2018062] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 11/16/2018] [Indexed: 12/14/2022]
Abstract
Background: Serpins are a superfamily of serine peptidase inhibitors that participate in the regulation of many physiological and cell peptidase-mediated processes in all organisms (e.g. in blood clotting, complement activation, fibrinolysis, inflammation, and programmed cell death). It was postulated that in the blood-feeding members of the monogenean family Diplozoidae, serpins could play an important role in the prevention of thrombus formation, activation of complement, inflammation in the host, and/or in the endogenous regulation of protein degradation. Results: In silico analysis showed that the DNA and primary protein structures of serpin from Eudiplozoon nipponicum (EnSerp1) are similar to other members of the serpin superfamily. The inhibitory potential of EnSerp1 on four physiologically-relevant serine peptidases (trypsin, factor Xa, kallikrein, and plasmin) was demonstrated and its presence in the worm’s excretory-secretory products (ESPs) was confirmed. Conclusion: EnSerp1 influences the activity of peptidases that play a role in blood coagulation, fibrinolysis, and complement activation. This inhibitory potential, together with the serpin’s presence in ESPs, suggests that it is likely involved in host-parasite interactions and could be one of the molecules involved in the control of feeding and prevention of inflammatory responses.
Collapse
Affiliation(s)
- Pavel Roudnický
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kamenice 753/5, 62500 Brno, Czech Republic
| | - Jiří Vorel
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kamenice 753/5, 62500 Brno, Czech Republic
| | - Jana Ilgová
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kamenice 753/5, 62500 Brno, Czech Republic
| | - Michal Benovics
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kamenice 753/5, 62500 Brno, Czech Republic
| | - Adam Norek
- Veterinary Research Institute, Hudcova 296/70, 62100 Brno, Czech Republic
| | - Lucie Jedličková
- Department of Parasitology, Faculty of Science, Charles University, Viničná 7, 12844 Prague 2, Czech Republic
| | - Libor Mikeš
- Department of Parasitology, Faculty of Science, Charles University, Viničná 7, 12844 Prague 2, Czech Republic
| | - David Potěšil
- Central European Institute of Technology, Masaryk University, Kamenice 753/5, 62500 Brno, Czech Republic
| | - Zbyněk Zdráhal
- Central European Institute of Technology, Masaryk University, Kamenice 753/5, 62500 Brno, Czech Republic - National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 753/5, 62500 Brno, Czech Republic
| | - Jan Dvořák
- School of Biological Sciences, Medical Biology Centre, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom - Department of Zoology and Fisheries, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences in Prague, Kamýcká 129, 16521 Prague, Czech Republic
| | - Milan Gelnar
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kamenice 753/5, 62500 Brno, Czech Republic
| | - Martin Kašný
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kamenice 753/5, 62500 Brno, Czech Republic - Department of Parasitology, Faculty of Science, Charles University, Viničná 7, 12844 Prague 2, Czech Republic
| |
Collapse
|
32
|
Chen C, Xu XF, Zhang RQ, Ma Y, Lv Y, Li JL, Shi Q, Xiao K, Sun J, Yang XD, Shi Q, Dong XP. Remarkable increases of α1-antichymotrypsin in brain tissues of rodents during prion infection. Prion 2018; 11:338-351. [PMID: 28956708 DOI: 10.1080/19336896.2017.1349590] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
α1-Antichymotrypsin (α1-ACT) belongs to a kind of acute-phase inflammatory protein. Recently, such protein has been proved exist in the amyloid deposits which is the hallmark of Alzheimer's disease, but limitedly reported in prion disease. To estimate the change of α1-ACT during prion infection, the levels of α1-ACT in the brain tissues of scrapie agents 263K-, 139A- and ME7-infected rodents were analyzed, respectively. Results shown that α1-ACT levels were significantly increased in the brain tissues of the three kinds of scrapie-infected rodents, displaying a time-dependent manner during prion infection. Immunohistochemistry assays revealed the increased α1-ACT mainly accumulated in some cerebral regions of rodents infected with prion, such as cortex, thalamus and cerebellum. Immunofluorescent assays illustrated ubiquitously localization of α1-ACT with GFAP positive astrocytes, Iba1-positive microglia and NeuN-positive neurons. Moreover, double-stained immunofluorescent assays and immunohistochemistry assays using series of brain slices demonstrated close morphological colocalization of α1-ACT signals with that of PrP and PrPSc in the brain slices of 263K-infected hamster. However, co-immunoprecipitation does not identify any detectable molecular interaction between the endogenous α1-ACT and PrP either in the brain homogenates of 263K-infected hamsters or in the lysates of prion-infected cultured cells. Our data here imply that brain α1-ACT is increased abnormally in various scrapie-infected rodent models. Direct molecular interaction between α1-ACT and PrP seems not to be essential for the morphological colocalization of those two proteins in the brain tissues of prion infection.
Collapse
Affiliation(s)
- Cao Chen
- a State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China
| | - Xiao-Feng Xu
- a State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China
| | - Ren-Qing Zhang
- a State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China.,b College of Life Science and Technology, Heilongjiang Bayi Agricultural University , Daqing , People's Republic of China
| | - Yue Ma
- a State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China
| | - Yan Lv
- a State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China
| | - Jian-Le Li
- a State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China
| | - Qiang Shi
- a State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China
| | - Kang Xiao
- a State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China
| | - Jing Sun
- a State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China
| | - Xiao-Dong Yang
- a State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China
| | - Qi Shi
- a State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China
| | - Xiao-Ping Dong
- a State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China
| |
Collapse
|
33
|
Molecular Mechanisms of Vaspin Action - From Adipose Tissue to Skin and Bone, from Blood Vessels to the Brain. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1111:159-188. [PMID: 30051323 DOI: 10.1007/5584_2018_241] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Visceral adipose tissue-derived serine protease inhibitor (vaspin) or SERPINA12 according to the serpin nomenclature was identified together with other genes and gene products that were specifically expressed or overexpressed in the intra-abdominal or visceral adipose tissue (AT) of the Otsuka Long-Evans Tokushima fatty rat. These rats spontaneously develop visceral obesity, insulin resistance, hyperinsulinemia and -glycemia, as well as hypertension and thus represent a well suited animal model of obesity and related metabolic disorders such as type 2 diabetes.The follow-up study reporting the cloning, expression and functional characterization of vaspin suggested the great and promising potential of this molecule to counteract obesity induced insulin resistance and inflammation and has since initiated over 300 publications, clinical and experimental, that have contributed to uncover the multifaceted functions and molecular mechanisms of vaspin action not only in the adipose, but in many different cells, tissues and organs. This review will give an update on mechanistic and structural aspects of vaspin with a focus on its serpin function, the physiology and regulation of vaspin expression, and will summarize the latest on vaspin function in various tissues such as the different adipose tissue depots as well as the vasculature, skin, bone and the brain.
Collapse
|
34
|
McLean TR, Rank MM, Smooker PM, Richardson SJ. Evolution of thyroid hormone distributor proteins. Mol Cell Endocrinol 2017; 459:43-52. [PMID: 28249735 DOI: 10.1016/j.mce.2017.02.038] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 02/24/2017] [Accepted: 02/24/2017] [Indexed: 01/08/2023]
Abstract
Thyroid hormones (THs) are evolutionarily old hormones, having effects on metabolism in bacteria, invertebrates and vertebrates. THs bind specific distributor proteins (THDPs) to ensure their efficient distribution through the blood and cerebrospinal fluid in vertebrates. Albumin is a THDP in the blood of all studied species of vertebrates, so may be the original vertebrate THDP. However, albumin has weak affinity for THs. Transthyretin (TTR) has been identified in the blood across different lineages in adults vs juveniles. TTR has intermediate affinity for THs. Thyroxine-binding globulin has only been identified in mammals and has high affinity for THs. Of these THDPs, TTR is the only one known to be synthesised in the brain and is involved in moving THs from the blood into the cerebrospinal fluid. We analysed the rates of evolution of these three THDPs: TTR has been most highly conserved and albumin has had the highest rate of divergence.
Collapse
Affiliation(s)
- Thomas R McLean
- School of Science, RMIT University, Bundoora, 3083 Victoria, Australia.
| | - Michelle M Rank
- School of Health and Biomedical Sciences, RMIT University, Bundoora, 3083 Victoria, Australia.
| | - Peter M Smooker
- School of Science, RMIT University, Bundoora, 3083 Victoria, Australia.
| | - Samantha J Richardson
- School of Health and Biomedical Sciences, RMIT University, Bundoora, 3083 Victoria, Australia.
| |
Collapse
|
35
|
Yazd NKK, Patel RR, Dellavalle RP, Dunnick CA. Genetic Risk Factors for Development of Atopic Dermatitis: a Systematic Review. CURRENT DERMATOLOGY REPORTS 2017. [DOI: 10.1007/s13671-017-0199-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
36
|
Calvo-Iglesias J, Pérez-Estévez D, González-Fernández Á. MSP22.8 is a protease inhibitor-like protein involved in shell mineralization in the edible mussel Mytilus galloprovincialis. FEBS Open Bio 2017; 7:1539-1556. [PMID: 28979842 PMCID: PMC5623705 DOI: 10.1002/2211-5463.12286] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 08/01/2017] [Accepted: 08/02/2017] [Indexed: 01/01/2023] Open
Abstract
The mussel shell protein 22.8 (MSP22.8) is recognized by a monoclonal antibody (M22.8) directed against larvae of the mussel Mytilus galloprovincialis. After being secreted by cells of the mantle-edge epithelium into the extrapallial (EP) space (the gap between the mantle and the shell), the protein is detected in the extrapallial fluid (EPF) and EP hemocytes and finally becomes part of the shell matrix framework in adult specimens of M. galloprovincialis. In the work described here, we show how MSP22.8 is detected in EPF samples from different species of mussels (M. galloprovincialis, Mytilus edulis, and Xenostrobus securis), and also as a shell matrix protein in M. galloprovincialis, Mytilus chilensis, and Perna canaliculus. A multistep purification strategy was employed to isolate the protein from the EPF, which was then analyzed by mass spectrometry in order to identify it. The results indicate that MSP22.8 is a serpin-like protein that has great similarity with the protease inhibitor-like protein-B1, reported previously for Mytilus coruscus. We suggest that MSP22.8 is part of a system offering protection from proteolysis during biomineralization and is also part of the innate immune system in mussels.
Collapse
Affiliation(s)
- Juan Calvo-Iglesias
- Immunology Biomedical Research Center (CINBIO) Centro Singular de investigación de Galicia Institute of Biomedical Research of Vigo (IBIV) University of Vigo Pontevedra Spain
| | | | - África González-Fernández
- Immunology Biomedical Research Center (CINBIO) Centro Singular de investigación de Galicia Institute of Biomedical Research of Vigo (IBIV) University of Vigo Pontevedra Spain
| |
Collapse
|
37
|
Nichols DB, De Martini W, Cottrell J. Poxviruses Utilize Multiple Strategies to Inhibit Apoptosis. Viruses 2017; 9:v9080215. [PMID: 28786952 PMCID: PMC5580472 DOI: 10.3390/v9080215] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 07/31/2017] [Accepted: 08/02/2017] [Indexed: 12/11/2022] Open
Abstract
Cells have multiple means to induce apoptosis in response to viral infection. Poxviruses must prevent activation of cellular apoptosis to ensure successful replication. These viruses devote a substantial portion of their genome to immune evasion. Many of these immune evasion products expressed during infection antagonize cellular apoptotic pathways. Poxvirus products target multiple points in both the extrinsic and intrinsic apoptotic pathways, thereby mitigating apoptosis during infection. Interestingly, recent evidence indicates that poxviruses also hijack cellular means of eliminating apoptotic bodies as a means to spread cell to cell through a process called apoptotic mimicry. Poxviruses are the causative agent of many human and veterinary diseases. Further, there is substantial interest in developing these viruses as vectors for a variety of uses including vaccine delivery and as oncolytic viruses to treat certain human cancers. Therefore, an understanding of the molecular mechanisms through which poxviruses regulate the cellular apoptotic pathways remains a top research priority. In this review, we consider anti-apoptotic strategies of poxviruses focusing on three relevant poxvirus genera: Orthopoxvirus, Molluscipoxvirus, and Leporipoxvirus. All three genera express multiple products to inhibit both extrinsic and intrinsic apoptotic pathways with many of these products required for virulence.
Collapse
Affiliation(s)
- Daniel Brian Nichols
- Department of Biological Sciences, Seton Hall University, South Orange, NJ 07039, USA.
| | - William De Martini
- Department of Biological Sciences, Seton Hall University, South Orange, NJ 07039, USA.
| | - Jessica Cottrell
- Department of Biological Sciences, Seton Hall University, South Orange, NJ 07039, USA.
| |
Collapse
|
38
|
Jendroszek A, Sønnichsen MS, Muñoz AC, Leyman K, Christensen A, Petersen SV, Wang T, Bendixen C, Panitz F, Andreasen PA, Jensen JK. Latency transition of plasminogen activator inhibitor type 1 is evolutionarily conserved. Thromb Haemost 2017; 117:1688-1699. [PMID: 28771275 DOI: 10.1160/th17-02-0102] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/11/2017] [Indexed: 02/04/2023]
Abstract
Plasminogen activator inhibitor type 1 (PAI-1) is a central regulator of fibrinolysis and tissue remodelling. PAI-1 belongs to the serpin superfamily and unlike other inhibitory serpins undergoes a spontaneous inactivation process under physiological conditions, termed latency transition. During latency transition the solvent exposed reactive centre loop is inserted into the central β-sheet A of the molecule, and is no longer accessible to reaction with the protease. More than three decades of research on mammalian PAI-1 has not been able to clarify the evolutionary advantage and physiological relevance of latency transition. In order to study the origin of PAI-1 latency transition, we produced PAI-1 from Spiny dogfish shark (Squalus acanthias) and African lungfish (Protopterus sp.), which represent central species in the evolution of vertebrates. Although human PAI-1 and the non-mammalian PAI-1 variants share only approximately 50 % sequence identity, our results showed that all tested PAI-1 variants undergo latency transition with a similar rate. Since the functional stability of PAI-1 can be greatly increased by substitution of few amino acid residues, we conclude that the ability to undergo latency transition must have been a specific selection criterion for the evolution of PAI-1. It appears that all PAI-1 molecules must harbour latency transition to fulfil their physiological function, stressing the importance to further pursue a complete understanding of this molecular phenomenon with possible implication to pharmacological intervention. Our results provide the next step in understanding how the complete role of this important protease inhibitor evolved along with the fibrinolytic system.
Collapse
|
39
|
Christiansen SC, Zuraw BL. Laboratory Approaches for Assessing Contact System Activation. Immunol Allergy Clin North Am 2017; 37:527-539. [DOI: 10.1016/j.iac.2017.04.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
40
|
Yang L, Rezaie AR. Characterization of Protein Z-Dependent Protease Inhibitor/Antithrombin Chimeras Provides Insight into the Serpin Specificity of Coagulation Proteases. ACS OMEGA 2017; 2:3276-3283. [PMID: 28782047 PMCID: PMC5537704 DOI: 10.1021/acsomega.7b00606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 06/28/2017] [Indexed: 06/07/2023]
Abstract
Protein Z (PZ)-dependent protease inhibitor (ZPI) and antithrombin (AT) are two physiological serpin inhibitors involved in the regulation of proteolytic activities of the blood coagulation cascade. ZPI has restricted protease specificity capable of inhibiting factors Xa (FXa) and XIa (FXIa) but exhibiting no reactivity with other coagulation proteases. Unlike ZPI, AT is a general inhibitor of all coagulation proteases and the only physiological inhibitor of factor IXa (FIXa). To understand the molecular determinants of protease specificity of the two serpins, we engineered two ZPI mutants in which the P12-P3' residues of the reactive center loop of ZPI were replaced with either P12-P3' or P12-P7' residues of AT (ZPI-ATP12-P3' and ZPI-ATP12-P7'). The reactivity of chimeras with FXa was improved ∼4-25-fold in the absence of PZ. Both chimeras inhibited FIXa with rate constants that were ∼2 orders of magnitude higher than the rate of the AT inhibition of the protease. PZ improved the reactivity of chimeras with FIXa by another 2 orders of magnitude, rendering the chimeras potent inhibitors of FIXa so that the PZ-mediated inhibitory activity of the ZPI-AT chimeras toward FIXa was ∼20-fold higher than that of the fondaparinux-catalyzed inhibition of FIXa by AT. Further studies revealed that the substitution of P1-Tyr of ZPI with an Arg is sufficient to convert the serpin to an effective inhibitor of FIXa. The potential therapeutic utility of the serpin chimeras as specific inhibitors of FIXa was diminished by findings that the chimeras function as effective substrates for other coagulation proteases.
Collapse
Affiliation(s)
- Likui Yang
- Cardiovascular
Biology Research Program, Oklahoma Medical
Research Foundation, Oklahoma
City, Oklahoma 73104, United States
| | - Alireza R. Rezaie
- Cardiovascular
Biology Research Program, Oklahoma Medical
Research Foundation, Oklahoma
City, Oklahoma 73104, United States
- Department
of Biochemistry and Molecular Biology, University
of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| |
Collapse
|
41
|
Goulas T, Ksiazek M, Garcia-Ferrer I, Sochaj-Gregorczyk AM, Waligorska I, Wasylewski M, Potempa J, Gomis-Rüth FX. A structure-derived snap-trap mechanism of a multispecific serpin from the dysbiotic human oral microbiome. J Biol Chem 2017; 292:10883-10898. [PMID: 28512127 PMCID: PMC5491774 DOI: 10.1074/jbc.m117.786533] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 05/04/2017] [Indexed: 10/19/2022] Open
Abstract
Enduring host-microbiome relationships are based on adaptive strategies within a particular ecological niche. Tannerella forsythia is a dysbiotic member of the human oral microbiome that inhabits periodontal pockets and contributes to chronic periodontitis. To counteract endopeptidases from the host or microbial competitors, T. forsythia possesses a serpin-type proteinase inhibitor called miropin. Although serpins from animals, plants, and viruses have been widely studied, those from prokaryotes have received only limited attention. Here we show that miropin uses the serpin-type suicidal mechanism. We found that, similar to a snap trap, the protein transits from a metastable native form to a relaxed triggered or induced form after cleavage of a reactive-site target bond in an exposed reactive-center loop. The prey peptidase becomes covalently attached to the inhibitor, is dragged 75 Å apart, and is irreversibly inhibited. This coincides with a large conformational rearrangement of miropin, which inserts the segment upstream of the cleavage site as an extra β-strand in a central β-sheet. Standard serpins possess a single target bond and inhibit selected endopeptidases of particular specificity and class. In contrast, miropin uniquely blocked many serine and cysteine endopeptidases of disparate architecture and substrate specificity owing to several potential target bonds within the reactive-center loop and to plasticity in accommodating extra β-strands of variable length. Phylogenetic studies revealed a patchy distribution of bacterial serpins incompatible with a vertical descent model. This finding suggests that miropin was acquired from the host through horizontal gene transfer, perhaps facilitated by the long and intimate association of T. forsythia with the human gingiva.
Collapse
Affiliation(s)
- Theodoros Goulas
- From the Proteolysis Lab, Structural Biology Unit, María de Maeztu Unit of Excellence, Molecular Biology Institute of Barcelona, Consejo Superior de Investigaciones Científicas, 08028 Barcelona, Spain
| | - Miroslaw Ksiazek
- the Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology and
- the Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky 40202
| | - Irene Garcia-Ferrer
- From the Proteolysis Lab, Structural Biology Unit, María de Maeztu Unit of Excellence, Molecular Biology Institute of Barcelona, Consejo Superior de Investigaciones Científicas, 08028 Barcelona, Spain
| | - Alicja M Sochaj-Gregorczyk
- the Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology and
- the Malopolska Centre of Biotechnology, Jagiellonian University, 30-387 Kraków, Poland, and
| | - Irena Waligorska
- the Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology and
| | - Marcin Wasylewski
- the Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology and
| | - Jan Potempa
- the Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology and
- the Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky 40202
| | - F Xavier Gomis-Rüth
- From the Proteolysis Lab, Structural Biology Unit, María de Maeztu Unit of Excellence, Molecular Biology Institute of Barcelona, Consejo Superior de Investigaciones Científicas, 08028 Barcelona, Spain,
| |
Collapse
|
42
|
Gollapalli K, Ghantasala S, Kumar S, Srivastava R, Rapole S, Moiyadi A, Epari S, Srivastava S. Subventricular zone involvement in Glioblastoma - A proteomic evaluation and clinicoradiological correlation. Sci Rep 2017; 7:1449. [PMID: 28469129 PMCID: PMC5431125 DOI: 10.1038/s41598-017-01202-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Accepted: 03/27/2017] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma multiforme (GBM), the most malignant of all gliomas is characterized by a high degree of heterogeneity and poor response to treatment. The sub-ventricular zone (SVZ) is the major site of neurogenesis in the brain and is rich in neural stem cells. Based on the proximity of the GBM tumors to the SVZ, the tumors can be further classified into SVZ+ and SVZ−. The tumors located in close contact with the SVZ are classified as SVZ+, while the tumors located distantly from the SVZ are classified as SVZ−. To gain an insight into the increased aggressiveness of SVZ+ over SVZ− tumors, we have used proteomics techniques like 2D-DIGE and LC-MS/MS to investigate any possible proteomic differences between the two subtypes. Serum proteomic analysis revealed significant alterations of various acute phase proteins and lipid carrying proteins, while tissue proteomic analysis revealed significant alterations in cytoskeletal, lipid binding, chaperone and cell cycle regulating proteins, which are already known to be associated with disease pathobiology. These findings provide cues to molecular basis behind increased aggressiveness of SVZ+ GBM tumors over SVZ− GBM tumors and plausible therapeutic targets to improve treatment modalities for these highly invasive tumors.
Collapse
Affiliation(s)
| | | | - Sachendra Kumar
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai, India
| | | | - Srikanth Rapole
- Proteomics Laboratory, National Centre for Cell Science, Ganeshkhind, Pune, India
| | - Aliasgar Moiyadi
- Advanced Centre for Treatment, Research and Education in Cancer (ACTREC) and Tata Memorial Hospital, Tata Memorial Centre, Kharghar, Navi Mumbai, Mumbai, India
| | - Sridhar Epari
- Advanced Centre for Treatment, Research and Education in Cancer (ACTREC) and Tata Memorial Hospital, Tata Memorial Centre, Kharghar, Navi Mumbai, Mumbai, India
| | | |
Collapse
|
43
|
Ali MF, Kaushik A, Kapil C, Gupta D, Jairajpuri MA. A hydrophobic patch surrounding Trp154 in human neuroserpin controls the helix F dynamics with implications in inhibition and aggregation. Sci Rep 2017; 7:42987. [PMID: 28230174 PMCID: PMC5322333 DOI: 10.1038/srep42987] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 01/17/2017] [Indexed: 01/23/2023] Open
Abstract
Neuroserpin (NS) mediated inhibition of tissue-type plasminogen activator (tPA) is important for brain development, synapse formation and memory. Aberrations in helix F and β-sheet A movement during inhibition can directly lead to epilepsy or dementia. Conserved W154 residue in a hydrophobic patch between helix F and β-sheet A is ideally placed to control their movement during inhibition. Molecular Dynamics (MD) simulation on wild type (WT) NS and its two variants (W154A and W154P) demonstrated partial deformation in helix F and conformational differences in strands 1A and 2A only in W154P. A fluorescence and Circular Dichroism (CD) analysis with purified W154 variants revealed a significant red-shift and an increase in α-helical content in W154P as compared to W154A and WT NS. Kinetics of tPA inhibition showed a decline in association rates (ka) for W154A as compared to WT NS with indication of complex formation. Appearance of cleaved without complex formation in W154P indicates that the variant acts as substrate due to conformational misfolding around helix F. Both the variants however showed increased rate of aggregation as compared to WT NS. The hydrophobic patch identified in this study may have importance in helix F dynamics of NS.
Collapse
Affiliation(s)
- Mohammad Farhan Ali
- Protein Conformation and Enzymology Lab, Department of Biosciences, Jamia Millia Islamia (A Central University), New Delhi-110025, India
| | - Abhinav Kaushik
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), Aruna Asaf Ali Marg, New Delhi-110067, India
| | - Charu Kapil
- Protein Conformation and Enzymology Lab, Department of Biosciences, Jamia Millia Islamia (A Central University), New Delhi-110025, India
| | - Dinesh Gupta
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), Aruna Asaf Ali Marg, New Delhi-110067, India
| | - Mohamad Aman Jairajpuri
- Protein Conformation and Enzymology Lab, Department of Biosciences, Jamia Millia Islamia (A Central University), New Delhi-110025, India
| |
Collapse
|
44
|
Abstract
Remarkable progress in understanding the pathophysiology and underlying mechanisms of hereditary angioedema has led to the development of effective treatment for this disorder. Progress in three separate areas has catalyzed our understanding of hereditary angioedema. The first is the recognition that HAE type I and type II result from a deficiency in the plasma level of functional C1 inhibitor. This observation has led to a detailed understanding of the SERPING1 mutations responsible for this deficiency as well as the molecular regulation of C1 inhibitor expression and function. The second is that the fundamental cause of swelling is enhanced contact system activation leading to increased generation of bradykinin. Substantial progress has been made in defining the parameters regulating bradykinin generation and catabolism as well as the receptors that transduce the biologic effects of kinins. The third is the understanding that tissue swelling in hereditary angioedema primarily involves the function of endothelial cell adherens junctions. This knowledge is driving increased attention to the role of endothelial biology in determining disease activity in hereditary angioedema. While there has been considerable progress made, large gaps still remain in our knowledge. Important areas that remain poorly understood include the factors that lead to very low plasma functional C1 inhibitor levels, the triggers of contact system activation in hereditary angioedema, and the role of the bradykinin B1 receptor. The phenotypic variability of hereditary angioedema has been extensively documented but never understood. The mechanisms discussed in this chapter likely contribute to this variability. Future progress in understanding these mechanisms should provide new means to improve the diagnosis and treatment of hereditary angioedema.
Collapse
|
45
|
Krishnan B, Hedstrom L, Hebert DN, Gierasch LM, Gershenson A. Expression and Purification of Active Recombinant Human Alpha-1 Antitrypsin (AAT) from Escherichia coli. Methods Mol Biol 2017; 1639:195-209. [PMID: 28752459 DOI: 10.1007/978-1-4939-7163-3_19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Well-established genetic manipulation procedures along with a fast doubling time, the ability to grow in inexpensive media, and easy scaleup make Escherichia coli (E. coli) a preferred recombinant protein expression platform. Human alpha-1 antitrypsin (AAT) and other serpins are easily expressed in E. coli despite their metastability and complicated topology. Serpins can be produced as soluble proteins or aggregates in inclusion bodies, and both forms can be purified to homogeneity. In this chapter, we describe an ion-exchange chromatography-based protocol that we have developed involving the use of two anion-exchange columns to purify untagged human AAT from E. coli. We also outline methods that can be used to determine the inhibitory activity of both AAT in cell lysates and purified AAT. Our protocol for the purification of bacterially expressed AAT yields pure and active protein at 6-7 mg/l culture.
Collapse
Affiliation(s)
- Beena Krishnan
- G.N. Ramachandran Protein Centre, CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, 160 036, India.
| | - Lizbeth Hedstrom
- Department of Biology, Brandeis University Waltham, Waltham, MA, USA.,Department of Chemistry, Brandeis University Waltham, Waltham, MA, USA
| | - Daniel N Hebert
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst, MA, USA
| | - Lila M Gierasch
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst, MA, USA.,Department of Chemistry, University of Massachusetts Amherst, Amherst, MA, USA
| | - Anne Gershenson
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst, MA, USA
| |
Collapse
|
46
|
Carrell RW, Read RJ. How serpins transport hormones and regulate their release. Semin Cell Dev Biol 2016; 62:133-141. [PMID: 28027946 DOI: 10.1016/j.semcdb.2016.12.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 12/09/2016] [Accepted: 12/15/2016] [Indexed: 12/24/2022]
Abstract
The adaptation of the serpin framework and its mechanism to perform diverse functions is epitomised in the hormone carriers of the blood. Thyroxine and the corticosteroids are transported bound in a 1:1 ratio on almost identical sites in the two homologous binding-globulins, TBG and CBG. Recent structural findings show an equilibrated, rather than on-and-off, release of the hormones from the carriers, reflecting small reversible movements of the hinge region of the reactive loop that modify the conformational flexibility of the underlying hormone-binding site. Consequently, contrary to previous concepts, the binding affinities of TBG and CBG are not fixed but can be allosterically modified to allow differential hormone delivery. Notably, the two carriers function like protein thermocouples with a surge in hormone release as body temperatures rise in fevers, and conversely a large diminution in free hormone levels at hibernation temperatures. By comparison angiotensinogen, the source of the angiotensin peptides that control blood pressure, does not appear to utilise the serpin mechanism. It has instead evolved a 63 residue terminal extension containing the buried angiotensin cleavage site, which on interaction moves into the active cleft of the renin. The conformational shift involved is critically linked by a labile disulphide bridge. The observation of changes in the redox status of this S-S bridge, in the hypertensive complication of pregnancy, pre-eclampsia, has opened an unexpected level of regulation at what is the initial stage in the control of blood pressure.
Collapse
Affiliation(s)
- Robin W Carrell
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK.
| | - Randy J Read
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| |
Collapse
|
47
|
Pippel J, Kuettner EB, Ulbricht D, Daberger J, Schultz S, Heiker JT, Sträter N. Crystal structure of cleaved vaspin (serpinA12). Biol Chem 2016; 397:111-23. [PMID: 26529565 DOI: 10.1515/hsz-2015-0229] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 10/28/2015] [Indexed: 11/15/2022]
Abstract
The adipokine vaspin (serpinA12) is mainly expressed in white adipose tissue and exhibits various beneficial effects on obesity-related processes. Kallikrein 7 is the only known target protease of vaspin and is inhibited by the classical serpin inhibitory mechanism involving a cleavage of the reactive center loop between P1 (M378) and P1' (E379). Here, we present the X-ray structure of vaspin, cleaved between M378 and E379. We provide a comprehensive analysis of differences between the uncleaved and cleaved forms in the shutter, breach, and hinge regions with relation to common molecular features underlying the serpin inhibitory mode. Furthermore, we point out differences towards other serpins and provide novel data underlining the remarkable stability of vaspin. We speculate that the previously reported FKGx1Wx2x3 motif in the breach region may play a decisive role in determining the reactive center loop configuration in the native vaspin state and might contribute to the high thermostability of vaspin. Thus, this structure may provide a basis for future mutational studies.
Collapse
|
48
|
The Microtubule Inhibitor Podofilox Inhibits an Early Entry Step of Human Cytomegalovirus. Viruses 2016; 8:v8100295. [PMID: 27783035 PMCID: PMC5086627 DOI: 10.3390/v8100295] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 10/11/2016] [Accepted: 10/14/2016] [Indexed: 12/20/2022] Open
Abstract
Human cytomegalovirus is a ubiquitous β-herpesvirus that infects many different cell types through an initial binding to cell surface receptors followed by a fusion event at the cell membrane or endocytic vesicle. A recent high-throughput screen to identify compounds that block a step prior to viral gene expression identified podofilox as a potent and nontoxic inhibitor. Time-of-addition studies in combination with quantitative-PCR analysis demonstrated that podofilox limits an early step of virus entry at the cell surface. Podofilox was also able to drastically reduce infection by herpes simplex 1, an α-herpesvirus with a very similar entry process to CMV. Podofilox caused a reduced maximal plateau inhibition of infection by viruses with single step binding processes prior to fusion-like Newcastle disease virus, Sendai virus, and influenza A virus or viruses that enter via endocytosis like vesicular stomatitis virus and a clinical-like strain of CMV. These results indicate that microtubules appear to be participating in the post-binding step of virus entry including the pre- and post-penetration events. Modulation of the plasma membrane is required to promote virus entry for herpesviruses, and that podofilox, unlike colchicine or nocodazole, is able to preferentially target microtubule networks at the plasma membrane.
Collapse
|
49
|
Henley D, Lightman S, Carrell R. Cortisol and CBG — Getting cortisol to the right place at the right time. Pharmacol Ther 2016; 166:128-35. [DOI: 10.1016/j.pharmthera.2016.06.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 06/24/2016] [Indexed: 02/06/2023]
|
50
|
Huang X, Zheng Y, Zhang F, Wei Z, Wang Y, Carrell RW, Read RJ, Chen GQ, Zhou A. Molecular Mechanism of Z α1-Antitrypsin Deficiency. J Biol Chem 2016; 291:15674-86. [PMID: 27246852 PMCID: PMC4957051 DOI: 10.1074/jbc.m116.727826] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Indexed: 12/14/2022] Open
Abstract
The Z mutation (E342K) of α1-antitrypsin (α1-AT), carried by 4% of Northern Europeans, predisposes to early onset of emphysema due to decreased functional α1-AT in the lung and to liver cirrhosis due to accumulation of polymers in hepatocytes. However, it remains unclear why the Z mutation causes intracellular polymerization of nascent Z α1-AT and why 15% of the expressed Z α1-AT is secreted into circulation as functional, but polymerogenic, monomers. Here, we solve the crystal structure of the Z-monomer and have engineered replacements to assess the conformational role of residue Glu-342 in α1-AT. The results reveal that Z α1-AT has a labile strand 5 of the central β-sheet A (s5A) with a consequent equilibrium between a native inhibitory conformation, as in its crystal structure here, and an aberrant conformation with s5A only partially incorporated into the central β-sheet. This aberrant conformation, induced by the loss of interactions from the Glu-342 side chain, explains why Z α1-AT is prone to polymerization and readily binds to a 6-mer peptide, and it supports that annealing of s5A into the central β-sheet is a crucial step in the serpins' metastable conformational formation. The demonstration that the aberrant conformation can be rectified through stabilization of the labile s5A by binding of a small molecule opens a potential therapeutic approach for Z α1-AT deficiency.
Collapse
Affiliation(s)
- Xin Huang
- From the Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine and University of Chinese Academy of Sciences, Shanghai 200025, China
| | - Ying Zheng
- the Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China, and
| | - Fei Zhang
- the Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China, and
| | - Zhenquan Wei
- the Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China, and
| | - Yugang Wang
- the Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China, and
| | - Robin W Carrell
- the Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, United Kingdom
| | - Randy J Read
- the Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, United Kingdom
| | - Guo-Qiang Chen
- From the Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine and University of Chinese Academy of Sciences, Shanghai 200025, China, the Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China, and
| | - Aiwu Zhou
- the Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China, and
| |
Collapse
|