1
|
Loch JI, Pieróg I, Imiołczyk B, Barciszewski J, Marsolais F, Gilski M, Jaskolski M. Unique double-helical packing of protein molecules in the crystal of potassium-independent L-asparaginase from common bean. Acta Crystallogr D Struct Biol 2025; 81:252-264. [PMID: 40243630 DOI: 10.1107/s205979832500292x] [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: 12/11/2024] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
Common bean (Phaseolus vulgaris) encodes three class 2 L-asparaginase enzymes: two potassium-dependent enzymes [PvAIII(K)-1 and PvAIII(K)-2] and a potassium-independent enzyme (PvAIII). Here, we present the crystal structure of PvAIII, which displays a rare P2 space-group symmetry and a unique pseudosymmetric 41-like double-helical packing. The asymmetric unit contains 32 protein chains (16 αβ units labeled A-P) organized into two right-handed coiled arrangements, each consisting of four PvAIII (αβ)2 dimers. Detailed analysis of the crystal structure revealed that this unusual packing originates from three factors: (i) the ability of the PvAIII molecules to form extended intermolecular β-sheets, a feature enabled by the PvAIII sequence and secondary structure, (ii) incomplete degradation of the flexible linker remaining at the C-terminus of α subunits of protein chain C after the autoproteolytic cleavage (maturation) of the PvAIII precursor and (iii) intermolecular entanglement between protein chains from the two helices to create `hydrogen-bond linchpins' that connect adjacent protein chains. The Km value of PvAIII for L-asparagine is approximately five times higher than for β-peptides, suggesting that the physiological role of PvAIII may be more related to the removal of toxic β-peptides than to basic L-asparagine metabolism. A comparison of the active sites of PvAIII and PvAIII(K)-1 shows that the proteins have nearly identical residues in the catalytic center, except for Thr219, which is unique to PvAIII. To test whether the residue type at position 219 affects the enzymatic activity of PvAIII, we designed and produced a T219S mutant. The kinetic parameters determined for L-asparagine hydrolysis indicate that the T/S residue type at position 219 does not affect the L-asparaginase activity of PvAIII.
Collapse
Affiliation(s)
- Joanna I Loch
- Department of Crystal Chemistry and Crystal Physics, Faculty of Chemistry, Jagiellonian University, Krakow, Poland
| | - Izabela Pieróg
- Department of Crystal Chemistry and Crystal Physics, Faculty of Chemistry, Jagiellonian University, Krakow, Poland
| | - Barbara Imiołczyk
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Jakub Barciszewski
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Frédéric Marsolais
- Genomics and Biotechnology, London Research and Development Centre, Agriculture and Agri-Food Canada, London, Ontario, Canada
| | - Mirosław Gilski
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Mariusz Jaskolski
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| |
Collapse
|
2
|
Wlodawer A, Dauter Z, Lubkowski J, Loch JI, Brzezinski D, Gilski M, Jaskolski M. Towards a dependable data set of structures for L-asparaginase research. Acta Crystallogr D Struct Biol 2024; 80:506-527. [PMID: 38935343 PMCID: PMC11220836 DOI: 10.1107/s2059798324005461] [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: 02/21/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
The Protein Data Bank (PDB) includes a carefully curated treasury of experimentally derived structural data on biological macromolecules and their various complexes. Such information is fundamental for a multitude of projects that involve large-scale data mining and/or detailed evaluation of individual structures of importance to chemistry, biology and, most of all, to medicine, where it provides the foundation for structure-based drug discovery. However, despite extensive validation mechanisms, it is almost inevitable that among the ∼215 000 entries there will occasionally be suboptimal or incorrect structure models. It is thus vital to apply careful verification procedures to those segments of the PDB that are of direct medicinal interest. Here, such an analysis was carried out for crystallographic models of L-asparaginases, enzymes that include approved drugs for the treatment of certain types of leukemia. The focus was on the adherence of the atomic coordinates to the rules of stereochemistry and their agreement with the experimental electron-density maps. Whereas the current clinical application of L-asparaginases is limited to two bacterial proteins and their chemical modifications, the field of investigations of such enzymes has expanded tremendously in recent years with the discovery of three entirely different structural classes and with numerous reports, not always quite reliable, of the anticancer properties of L-asparaginases of different origins.
Collapse
Affiliation(s)
- Alexander Wlodawer
- Center for Structural Biology, Center for Cancer ResearchNational Cancer InstituteMarylandUSA
| | - Zbigniew Dauter
- Center for Structural Biology, Center for Cancer ResearchNational Cancer InstituteMarylandUSA
| | - Jacek Lubkowski
- Center for Structural Biology, Center for Cancer ResearchNational Cancer InstituteMarylandUSA
| | - Joanna I. Loch
- Department of Crystal Chemistry and Crystal Physics, Faculty of ChemistryJagiellonian UniversityCracowPoland
| | - Dariusz Brzezinski
- Institute of Computing SciencePoznan University of TechnologyPoznanPoland
| | - Miroslaw Gilski
- Institute of Bioorganic ChemistryPolish Academy of SciencesPoznanPoland
- Department of Crystallography, Faculty of Chemistry, Adam Mickiewicz University, Poznan, Poland
| | - Mariusz Jaskolski
- Institute of Bioorganic ChemistryPolish Academy of SciencesPoznanPoland
- Department of Crystallography, Faculty of Chemistry, Adam Mickiewicz University, Poznan, Poland
| |
Collapse
|
3
|
Darvishi F, Beiranvand E, Kalhor H, Shahbazi B, Mafakher L. Homology modeling and molecular docking studies to decrease glutamine affinity of Yarrowia lipolytica L-asparaginase. Int J Biol Macromol 2024; 263:130312. [PMID: 38403216 DOI: 10.1016/j.ijbiomac.2024.130312] [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: 10/13/2023] [Revised: 01/10/2024] [Accepted: 02/18/2024] [Indexed: 02/27/2024]
Abstract
L-Asparaginase is a key component in the treatment of leukemias and lymphomas. However, the glutamine affinity of this therapeutic enzyme is an off-target activity that causes several side effects. The modeling and molecular docking study of Yarrowia lipolytica L-asparaginase (YL-ASNase) to reduce its l-glutamine affinity and increase its stability was the aim of this study. Protein-ligand interactions of wild-type and different mutants of YL-ASNase against L-asparagine compared to l-glutamine were assessed using AutoDock Vina tools because the crystal structure of YL-ASNase does not exist in the protein data banks. The results showed that three mutants, T171S, T171S-N60A, and T171A-T223A, caused a considerable increase in L-asparagine affinity and a decrease in l-glutamine affinity as compared to the wild-type and other mutants. Then, molecular dynamics simulation and MM/GBSA free energy were applied to assess the stability of protein structure and its interaction with ligands. The three mutated proteins, especially T171S-N60A, had higher stability and interactions with L-asparagine than l-glutamine in comparison with the wild-type. The YL-ASNase mutants could be introduced as appropriate therapeutic candidates that might cause lower side effects. However, the functional properties of these mutated enzymes need to be confirmed by genetic manipulation and in vitro and in vivo studies.
Collapse
Affiliation(s)
- Farshad Darvishi
- Department of Microbiology, Faculty of Biological Sciences, Alzahra University, Tehran, Iran; Research Center for Applied Microbiology and Microbial Biotechnology (CAMB), Alzahra University, Tehran, Iran.
| | - Elham Beiranvand
- Department of Microbiology, Faculty of Biological Sciences, Alzahra University, Tehran, Iran.
| | - Hourieh Kalhor
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Behzad Shahbazi
- School of Pharmacy, Semnan University of Medical Sciences, Semnan, Iran
| | - Ladan Mafakher
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
4
|
Sliwiak J, Worsztynowicz P, Pokrywka K, Loch JI, Grzechowiak M, Jaskolski M. Biochemical characterization of L-asparaginase isoforms from Rhizobium etli-the boosting effect of zinc. Front Chem 2024; 12:1373312. [PMID: 38456185 PMCID: PMC10917881 DOI: 10.3389/fchem.2024.1373312] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 02/09/2024] [Indexed: 03/09/2024] Open
Abstract
L-Asparaginases, divided into three structural Classes, catalyze the hydrolysis of L-asparagine to L-aspartic acid and ammonia. The members of Class 3, ReAIV and ReAV, encoded in the genome of the nitrogen fixing Rhizobium etli, have the same fold, active site, and quaternary structure, despite low sequence identity. In the present work we examined the biochemical consequences of this difference. ReAIV is almost twice as efficient as ReAV in asparagine hydrolysis at 37°C, with the kinetic KM, kcat parameters (measured in optimal buffering agent) of 1.5 mM, 770 s-1 and 2.1 mM, 603 s-1, respectively. The activity of ReAIV has a temperature optimum at 45°C-55°C, whereas the activity of ReAV, after reaching its optimum at 37°C, decreases dramatically at 45°C. The activity of both isoforms is boosted by 32 or 56%, by low and optimal concentration of zinc, which is bound three times more strongly by ReAIV then by ReAV, as reflected by the KD values of 1.2 and 3.3 μM, respectively. We also demonstrate that perturbation of zinc binding by Lys→Ala point mutagenesis drastically decreases the enzyme activity but also changes the mode of response to zinc. We also examined the impact of different divalent cations on the activity, kinetics, and stability of both isoforms. It appeared that Ni2+, Cu2+, Hg2+, and Cd2+ have the potential to inhibit both isoforms in the following order (from the strongest to weakest inhibitors) Hg2+ > Cu2+ > Cd2+ > Ni2+. ReAIV is more sensitive to Cu2+ and Cd2+, while ReAV is more sensitive to Hg2+ and Ni2+, as revealed by IC50 values, melting scans, and influence on substrate specificity. Low concentration of Cd2+ improves substrate specificity of both isoforms, suggesting its role in substrate recognition. The same observation was made for Hg2+ in the case of ReAIV. The activity of the ReAV isoform is less sensitive to Cl- anions, as reflected by the IC50 value for NaCl, which is eightfold higher for ReAV relative to ReAIV. The uncovered complementary properties of the two isoforms help us better understand the inducibility of the ReAV enzyme.
Collapse
Affiliation(s)
- Joanna Sliwiak
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | | | - Kinga Pokrywka
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Joanna I. Loch
- Department of Crystal Chemistry and Crystal Physics, Faculty of Chemistry, Jagiellonian University, Krakow, Poland
| | - Marta Grzechowiak
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Mariusz Jaskolski
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
- Department of Crystallography, Faculty of Chemistry, Adam Mickiewicz University, Poznan, Poland
| |
Collapse
|
5
|
Comparative structural and kinetic study for development of a novel candidate L-asparaginase based pharmaceutical. Biochem Eng J 2023. [DOI: 10.1016/j.bej.2023.108806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
6
|
Sharma A, Kaushik V, Goel M. Insights into the Distribution and Functional Properties of l-Asparaginase in the Archaeal Domain and Characterization of Picrophilus torridus Asparaginase Belonging to the Novel Family Asp2like1. ACS OMEGA 2022; 7:40750-40765. [PMID: 36406543 PMCID: PMC9670692 DOI: 10.1021/acsomega.2c01127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
l-Asparaginase catalyzes the hydrolysis of l-asparagine to aspartic acid and ammonia and is used in the medical and food industries. In this investigation, from the proteomes of 176 archaeal organisms (with completely sequenced genomes), 116 homologs of l-asparaginase were obtained from 86 archaeal organisms segregated into Asp1, Asp2, IaaA, Asp2like1, and Asp2like2 families based on the conserved domain. The similarities and differences in the structure of selected representatives from each family are discussed. From the two novel archaeal l-asparaginase families Asp2like1 and Asp2like2, a representative of Asp2like1 family Picrophilus torridus asparaginase (PtAsp2like1) was characterized in detail to find its suitability in therapeutics. PtAsp2like1 was a glutaminase-free asparaginase that showed the optimum activity at 80 °C and pH 10.0. The Km of PtAsp2like1 toward substrate l-asparagine was 11.69 mM. This study demonstrates the improved mapping of asparaginases in the archaeal domain, facilitating future focused research on archaeal asparaginases for therapeutic applications.
Collapse
|
7
|
El-Fakharany E, Orabi H, Abdelkhalek E, Sidkey N. Purification and biotechnological applications of L-asparaginase from newly isolated Bacillus halotolerans OHEM18 as antitumor and antioxidant agent. J Biomol Struct Dyn 2022; 40:3837-3849. [PMID: 33228468 DOI: 10.1080/07391102.2020.1851300] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/10/2020] [Indexed: 10/22/2022]
Abstract
In the present study, a new bacterial strain, Bacillus halotolerans OHEM18 was significantly found to produce extracellular L-asparaginase. L-asparaginase was purified using ammonium sulfate precipitation and QFF column to 3.84-fold with specific activity of 215.33 U/mg and its molecular mass was assessed to be 41.5 kDa. Maximum enzyme activity was determined at pH 8.2 and 40 °C and with retaining 70% of its activity after incubation for 1 h at 50 °C. Km and Vmax values were determined to be 0.0047 M and 92.74, respectively. Cytotoxicity test indicated a significant safety of L-asparaginase on Vero cells with selectivity against leukemia, breast cancer and hepatoma cells. NFS-60 cells was the most sensitive tumor cells to L-asparaginase with IC50 of 10.29 µg/ml and selectivity index of 30.61. This selectivity was recognized to be an apoptosis-dependent mechanism proven via cell cycle arrest in sub-G1 phase and fragmentation of genomic DNA. L-asparaginase showed antioxidant activity against both DPPH and ABTS radicals with IC50 values of 64.07 and 177.1 mg/ml, respectively. These competitive advantage of bacterial L-asparaginase over than other sources is that it might be produced in large amounts through production in large-scale biofermenters, which decreases costs, besides having a sustainable bacterial source. Our findings established that the potent cytotoxic effect of L-asparaginase isolated from B. halotolerans OHEM18 may be a promise candidate for further medicinal applications as an antioxidant and antitumor drug.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Esmail El-Fakharany
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), Alexandria, Egypt
| | - Hanaa Orabi
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), Alexandria, Egypt
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | - Eman Abdelkhalek
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | - Nagwa Sidkey
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
8
|
Dumina M, Zhgun A, Pokrovskaya M, Aleksandrova S, Zhdanov D, Sokolov N, El’darov M. Highly Active Thermophilic L-Asparaginase from Melioribacter roseus Represents a Novel Large Group of Type II Bacterial L-Asparaginases from Chlorobi-Ignavibacteriae-Bacteroidetes Clade. Int J Mol Sci 2021; 22:13632. [PMID: 34948436 PMCID: PMC8709496 DOI: 10.3390/ijms222413632] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 02/07/2023] Open
Abstract
L-asparaginase (L-ASNase) is a biotechnologically relevant enzyme for the pharmaceutical, biosensor and food industries. Efforts to discover new promising L-ASNases for different fields of biotechnology have turned this group of enzymes into a growing family with amazing diversity. Here, we report that thermophile Melioribacter roseus from Ignavibacteriae of the Bacteroidetes/Chlorobi group possesses two L-ASNases-bacterial type II (MrAII) and plant-type (MrAIII). The current study is focused on a novel L-ASNase MrAII that was expressed in Escherichia coli, purified and characterized. The enzyme is optimally active at 70 °C and pH 9.3, with a high L-asparaginase activity of 1530 U/mg and L-glutaminase activity ~19% of the activity compared with L-asparagine. The kinetic parameters KM and Vmax for the enzyme were 1.4 mM and 5573 µM/min, respectively. The change in MrAII activity was not significant in the presence of 10 mM Ni2+, Mg2+ or EDTA, but increased with the addition of Cu2+ and Ca2+ by 56% and 77%, respectively, and was completely inhibited by Zn2+, Fe3+ or urea solutions 2-8 M. MrAII displays differential cytotoxic activity: cancer cell lines K562, Jurkat, LnCap, and SCOV-3 were more sensitive to MrAII treatment, compared with normal cells. MrAII represents the first described enzyme of a large group of uncharacterized counterparts from the Chlorobi-Ignavibacteriae-Bacteroidetes clade.
Collapse
Affiliation(s)
- Maria Dumina
- Group of Fungal Genetic Engineering, Federal Research Center “Fundamentals of Biotechnology” of the Russian Academy of Sciences, 117312 Moscow, Russia;
| | - Alexander Zhgun
- Group of Fungal Genetic Engineering, Federal Research Center “Fundamentals of Biotechnology” of the Russian Academy of Sciences, 117312 Moscow, Russia;
| | - Marina Pokrovskaya
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, 119121 Moscow, Russia; (M.P.); (S.A.); (D.Z.); (N.S.)
| | - Svetlana Aleksandrova
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, 119121 Moscow, Russia; (M.P.); (S.A.); (D.Z.); (N.S.)
| | - Dmitry Zhdanov
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, 119121 Moscow, Russia; (M.P.); (S.A.); (D.Z.); (N.S.)
| | - Nikolay Sokolov
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, 119121 Moscow, Russia; (M.P.); (S.A.); (D.Z.); (N.S.)
| | - Michael El’darov
- Group of Fungal Genetic Engineering, Federal Research Center “Fundamentals of Biotechnology” of the Russian Academy of Sciences, 117312 Moscow, Russia;
| |
Collapse
|
9
|
Lee JK, Park HJ, Cha SJ, Kwon SJ, Park JH. Effect of pyroligneous acid on soil urease, amidase, and nitrogen use efficiency by Chinese cabbage (Brassica campestris var. Pekinensis). ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 291:118132. [PMID: 34536645 DOI: 10.1016/j.envpol.2021.118132] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 08/05/2021] [Accepted: 09/06/2021] [Indexed: 06/13/2023]
Abstract
Urea is one of the most commonly used nitrogen fertilizers in agricultural soil and is easily decomposed by soil urease resulting in ammonium release. The produced ammonium can be volatilized or converted to nitrate, which is susceptible to leaching, leading to groundwater contamination unless used by plants. Hence, it is important to control the release of nitrogen from the urea. Pyroligneous acid inhibited the urease activity and decreased ammonium release up to 80% compared to the control. Amidase including asparaginase and glutaminase is an enzyme that catalyzes hydrolysis of amide group, similar to urease. Therefore, the effect of pyroligneous acid on the inhibition of soil amidase was also tested and the results showed that pyroligneous acid competitively inhibited asparaginase while glutaminase was not inhibited. However, inhibitory effect of pyroligneous acid on asparaginase was negligible compared to the urease. The application of pyroligneous acid with a smaller amount of urea for controlled nitrogen release during Chinese cabbage growth showed that dry biomass and nutrient contents of Chinese cabbage were similar to the case of the conventional urea application. The nitrogen utilization efficiency (NUE) was highest for 33% less amount of urea supply with pyroligneous acid (2.21) compared to conventional treatment (1.81). Consequently, the use of pyroligneous acid with urea enhances nitrogen use efficiency while also protecting environments from non-point source contamination.
Collapse
Affiliation(s)
- Joo Kyung Lee
- Chungbuk National University, Cheongju, Chungbuk, 28644, South Korea
| | - Hyun Jun Park
- Soil Research Institute, Prumbio Co. Ltd, South Korea
| | - Seung Ju Cha
- Chungbuk National University, Cheongju, Chungbuk, 28644, South Korea
| | - Seon Ju Kwon
- Chungbuk National University, Cheongju, Chungbuk, 28644, South Korea
| | - Jin Hee Park
- Chungbuk National University, Cheongju, Chungbuk, 28644, South Korea.
| |
Collapse
|
10
|
Loch JI, Jaskolski M. Structural and biophysical aspects of l-asparaginases: a growing family with amazing diversity. IUCRJ 2021; 8:514-531. [PMID: 34258001 PMCID: PMC8256714 DOI: 10.1107/s2052252521006011] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 06/09/2021] [Indexed: 06/13/2023]
Abstract
l-Asparaginases have remained an intriguing research topic since their discovery ∼120 years ago, especially after their introduction in the 1960s as very efficient antileukemic drugs. In addition to bacterial asparaginases, which are still used to treat childhood leukemia, enzymes of plant and mammalian origin are now also known. They have all been structurally characterized by crystallography, in some cases at outstanding resolution. The structural data have also shed light on the mechanistic details of these deceptively simple enzymes. Yet, despite all this progress, no better therapeutic agents have been found to beat bacterial asparaginases. However, a new option might arise with the discovery of yet another type of asparaginase, those from symbiotic nitrogen-fixing Rhizobia, and with progress in the protein engineering of enzymes with desired properties. This review surveys the field of structural biology of l-asparaginases, focusing on the mechanistic aspects of the well established types and speculating about the potential of the new members of this amazingly diversified family.
Collapse
Affiliation(s)
- Joanna I. Loch
- Department of Crystal Chemistry and Crystal Physics, Faculty of Chemistry, Jagiellonian University, Cracow, Poland
| | - Mariusz Jaskolski
- Department of Crystallography, Faculty of Chemistry, A. Mickiewicz University, Poznan, Poland
- Center for Biocrystallographic Research, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| |
Collapse
|
11
|
Ran T, Jiao L, Wang W, Chen J, Chi H, Lu Z, Zhang C, Xu D, Lu F. Structures of l-asparaginase from Bacillus licheniformis Reveal an Essential Residue for its Substrate Stereoselectivity. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:223-231. [PMID: 33371681 DOI: 10.1021/acs.jafc.0c06609] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
l-Asparaginase, which catalyzes the hydrolysis of l-asparagine, is an important enzyme in both the clinical and food industry. Exploration of efficient l-asparaginase with high substrate specificity, especially high chiral selectivity, is essential for extending its use. Herein, various crystal structures of type I l-asparaginase from Bacillus licheniformis (BlAsnase) have been resolved, and we found that there are two additional tyrosines in BlAsnase, contributing to the binding and catalysis of d-asparagine. Strikingly, the substitution of Tyr278 with methionine impaired the interaction with d-asparagine via water molecules due to the small hydrophobic side chain of methionine, which forced the ligand to the deep side of the active site toward the catalytic residues and thus resulted in the loss of hydrolyzing function. Our investigation of the substrate recognition mechanism of BlAsnase is significant for both a better understanding of l-asparaginase and its rational design to achieve high specificity for clinical and industrial applications.
Collapse
Affiliation(s)
- Tingting Ran
- College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, China
| | - Linshu Jiao
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Weiwu Wang
- College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, China
| | - Juhua Chen
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Huibing Chi
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhaoxin Lu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Chong Zhang
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Dongqing Xu
- College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, China
| | - Fengxia Lu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
12
|
Saeed H, Hemida A, El-Nikhely N, Abdel-Fattah M, Shalaby M, Hussein A, Eldoksh A, Ataya F, Aly N, Labrou N, Nematalla H. Highly efficient Pyrococcus furiosus recombinant L-asparaginase with no glutaminase activity: Expression, purification, functional characterization, and cytotoxicity on THP-1, A549 and Caco-2 cell lines. Int J Biol Macromol 2020; 156:812-828. [PMID: 32311402 DOI: 10.1016/j.ijbiomac.2020.04.080] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/10/2020] [Accepted: 04/11/2020] [Indexed: 02/06/2023]
Abstract
L-Asparaginase (L-ASNase EC 3.5.1.1) is considered as an important biopharmaceutical drug enzyme in the treatment of childhood acute lymphoblastic leukemia (ALL). In the present study, Pyrococcus furiosus L-ASNase gene was cloned into pET26b (+), expressed in E. coli BL21(DE3) pLysS, and purified to homogeneity using Ni2+ chelated Fast Flow Sepharose resin with 5.7 purification fold and 23.9% recovery. The purified enzyme exhibited a molecular weight of ~33,660 Da on SDS-PAGE and showed maximal activity at 50 °C and pH 8.0. It retained 98.3% and 60.7% initial activity after 60 min at 37 °C and 50 °C, respectively. The recombinant enzyme showed highest substrate specificity towards L-ASNase substrate, while no detectable specificity was observed for l-glutamine, urea, and acrylamide at 10 mM concentration. The Km and Vmax of the purified recombinant enzyme as calculated using Lineweaver-Burk plot were determined to be 1.623 mM and 105 μmol min-1 mg-1, respectively. Human leukemia cell line THP-1 treated with recombinant L-ASNase showed significant morphological changes, and the IC50 of the purified enzyme was found to be 0.8 IU. Moreover, the purified recombinant L-ASNase induced cytotoxic effects on lung adenocarcinoma A549 and colorectal adenocarcinoma Caco-2 cell lines with IC50 of 1.78 IU and 30 IU, respectively.
Collapse
Affiliation(s)
- Hesham Saeed
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt.
| | - Asmaa Hemida
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Nefertiti El-Nikhely
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Manal Abdel-Fattah
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Manal Shalaby
- Genetic Engineering and Biotechnology Research Institute (GEBRI), City for Scientific Research and Technology Applications, New Borg Al-Arab City, Alexandria, Egypt
| | - Ahmed Hussein
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Ahmad Eldoksh
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Farid Ataya
- Biochemistry Department, College of Science, King Saud University, Bld. 5, Lab AA10, P.O. Box: 2454, Riyadh, Saudi Arabia; National Research Centre, 33 El-Bohouth St. (former El-Tahrir St.), Dokki, Giza 12622, Egypt
| | - Nihal Aly
- Botany and Microbiology Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Nikolaos Labrou
- Laboratory of Enzyme Technology, Department of Biotechnology, School of Food, Biotechnology and Development, Agricultural University of Athens, 75 lera Odos Street, Athens GR-11855, Greece
| | - Hisham Nematalla
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Damanhur University, Damnhour, Egypt
| |
Collapse
|
13
|
Gul A, Hussain G, Iqbal A, Rao AQ, Din SU, Yasmeen A, Shahid N, Ahad A, Latif A, Azam S, Samiullah TR, Hassan S, Shahid AA, Husnain T. Constitutive expression of Asparaginase in Gossypium hirsutum triggers insecticidal activity against Bemisia tabaci. Sci Rep 2020; 10:8958. [PMID: 32488033 PMCID: PMC7265412 DOI: 10.1038/s41598-020-65249-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 04/16/2020] [Indexed: 02/06/2023] Open
Abstract
Whitefly infestation of cotton crop imparts enormous damage to cotton yield by severely affecting plant health, vigour and transmitting Cotton Leaf Curl Virus (CLCuV). Genetic modification of cotton helps to overcome both the direct whitefly infestation as well as CLCuV based cotton yield losses. We have constitutively overexpressed asparaginase (ZmASN) gene in Gossypium hirsutum to overcome the cotton yield losses imparted by whitefly infestation. We achieved 2.54% transformation efficiency in CIM-482 by Agrobacterium-mediated shoot apex transformation method. The relative qRT-PCR revealed 40-fold higher transcripts of asparaginase in transgenic cotton line vs. non-transgenic cotton lines. Metabolic analysis showed higher contents of aspartic acid and glutamic acid in seeds and phloem sap of the transgenic cotton lines. Phenotypically, the transgenic cotton lines showed vigorous growth and height, greater number of bolls, and yield. Among six representative transgenic cotton lines, line 14 had higher photosynthetic rate, stomatal conductance, smooth fiber surface, increased fiber convolutions (SEM analysis) and 95% whitefly mortality as compared to non-transgenic cotton line. The gene integration analysis by fluorescence in situ hybridization showed single copy gene integration at chromosome number 1. Collectively, asparaginase gene demonstrated potential to control whitefly infestation, post-infestation damages and improve cotton plant health and yield: a pre-requisite for farmer's community.
Collapse
Affiliation(s)
- Ambreen Gul
- Centre of Excellence in Molecular Biology, University of the Punjab, 87-West Canal Bank Road, Lahore, 53700, Pakistan
- Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Ghulam Hussain
- Centre of Excellence in Molecular Biology, University of the Punjab, 87-West Canal Bank Road, Lahore, 53700, Pakistan
| | - Adnan Iqbal
- Centre of Excellence in Molecular Biology, University of the Punjab, 87-West Canal Bank Road, Lahore, 53700, Pakistan
| | - Abdul Qayyum Rao
- Centre of Excellence in Molecular Biology, University of the Punjab, 87-West Canal Bank Road, Lahore, 53700, Pakistan.
| | - Salah Ud Din
- Centre of Excellence in Molecular Biology, University of the Punjab, 87-West Canal Bank Road, Lahore, 53700, Pakistan
| | - Aneela Yasmeen
- Centre of Excellence in Molecular Biology, University of the Punjab, 87-West Canal Bank Road, Lahore, 53700, Pakistan
| | - Naila Shahid
- Centre of Excellence in Molecular Biology, University of the Punjab, 87-West Canal Bank Road, Lahore, 53700, Pakistan
| | - Ammara Ahad
- Centre of Excellence in Molecular Biology, University of the Punjab, 87-West Canal Bank Road, Lahore, 53700, Pakistan
| | - Ayesha Latif
- Centre of Excellence in Molecular Biology, University of the Punjab, 87-West Canal Bank Road, Lahore, 53700, Pakistan
| | - Saira Azam
- Centre of Excellence in Molecular Biology, University of the Punjab, 87-West Canal Bank Road, Lahore, 53700, Pakistan
| | - Tahir Rehman Samiullah
- Centre of Excellence in Molecular Biology, University of the Punjab, 87-West Canal Bank Road, Lahore, 53700, Pakistan
| | - Samina Hassan
- Centre of Excellence in Molecular Biology, University of the Punjab, 87-West Canal Bank Road, Lahore, 53700, Pakistan
- Kinnaird College for Women University, Lahore, Pakistan
| | - Ahmad Ali Shahid
- Centre of Excellence in Molecular Biology, University of the Punjab, 87-West Canal Bank Road, Lahore, 53700, Pakistan
| | - Tayyab Husnain
- Centre of Excellence in Molecular Biology, University of the Punjab, 87-West Canal Bank Road, Lahore, 53700, Pakistan
| |
Collapse
|
14
|
Chand S, Mahajan RV, Prasad JP, Sahoo DK, Mihooliya KN, Dhar MS, Sharma G. A comprehensive review on microbial l-asparaginase: Bioprocessing, characterization, and industrial applications. Biotechnol Appl Biochem 2020; 67:619-647. [PMID: 31954377 DOI: 10.1002/bab.1888] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 01/06/2020] [Indexed: 12/14/2022]
Abstract
l-Asparaginase (E.C.3.5.1.1.) is a vital enzyme that hydrolyzes l-asparagine to l-aspartic acid and ammonia. This property of l-asparaginase inhibits the protein synthesis in cancer cells, making l-asparaginase a mainstay of pediatric chemotherapy practices to treat acute lymphoblastic leukemia (ALL) patients. l-Asparaginase is also recognized as one of the important food processing agent. The removal of asparagine by l-asparaginase leads to the reduction of acrylamide formation in fried food items. l-Asparaginase is produced by various organisms including animals, plants, and microorganisms, however, only microorganisms that produce a substantial amount of this enzyme are of commercial significance. The commercial l-asparaginase for healthcare applications is chiefly derived from Escherichia coli and Erwinia chrysanthemi. A high rate of hypersensitivity and adverse reactions limits the long-term clinical use of l-asparaginase. Present review provides thorough information on microbial l-asparaginase bioprocess optimization including submerged fermentation and solid-state fermentation for l-asparaginase production, downstream purification, its characterization, and issues related to the clinical application including toxicity and hypersensitivity. Here, we have highlighted the bioprocess techniques that can produce improved and economically viable yields of l-asparaginase from promising microbial sources in the current scenario where there is an urgent need for alternate l-asparaginase with less adverse effects.
Collapse
Affiliation(s)
- Subhash Chand
- National Institute of Biologicals (Ministry of Health & Family Welfare, Government of India), Noida, Uttar Pradesh, India.,Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Richi V Mahajan
- National Institute of Biologicals (Ministry of Health & Family Welfare, Government of India), Noida, Uttar Pradesh, India
| | - Jai Prakash Prasad
- National Institute of Biologicals (Ministry of Health & Family Welfare, Government of India), Noida, Uttar Pradesh, India
| | - Debendra K Sahoo
- Council of Scientific and Industrial Research (CSIR)-Institute of Microbial Technology, Chandigarh, India
| | - Kanti Nandan Mihooliya
- Council of Scientific and Industrial Research (CSIR)-Institute of Microbial Technology, Chandigarh, India
| | - Mahesh S Dhar
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Girish Sharma
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India.,Amity Centre for Cancer Epidemiology & Cancer Research, Amity University, Noida, Uttar Pradesh, India
| |
Collapse
|
15
|
Molecular docking study of L-Asparaginase I from Vibrio campbellii in the treatment of acute lymphoblastic leukemia (ALL). EUROBIOTECH JOURNAL 2020. [DOI: 10.2478/ebtj-2020-0002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Abstract
The potential use of asparaginases has gained tremendous significance in the treatment of acute lymphoblastic leukemia (ALL). Earlier studies suggest L-asparaginases (L-ASP) extracted from Escherichia coli and Erwinia aroideae regulates L-asparagine (L-Asn) from the circulating blood. Prolonged exposure to these enzymes may lead to hypersensitivity reactions. So, it is important to find novel asparaginases with anti-cancer properties. The three-dimensional structure of L-ASP I from Vibrio campbellii was determined by homology modeling using EasyModeller v.4.0. The structure was validated with quality indexing tools and was deposited in Protein Model DataBase. Molecular docking was performed between L-ASP I and ligand substrate L-Asn to study enzyme-substrate interactions. Qualitative and quantitative analysis of L-ASP I enzyme was found to be reliable and stable with a significant protein quality factor (LG score: 7.129). The enzyme is a dimer, belongs to α/β class of proteins. The active sites comprises of N-glycosylation site and a catalytic triad (T14-S117-D92). The binding energy of the docked complex was calculated to be -7.45 kcal/mol. The amino acid T14 identified as a primary nucleophile essential for catalytic reaction. The enzyme L-ASP I of V. campbellii provides a detailed view of structure and functional aspects with ligand substrate L-Asn. This in silico investigation has explicitly demonstrated for the first time that cytosolic L-ASP Type I of V. campbellii to have a catalytic triad which was attributed only to periplasmic L-ASP Type II. Thus, L-ASP I can serve as anti-leukemic agent in the treatment, management and control of ALL.
Collapse
|
16
|
In silico analysis of codon usage and rare codon clusters in the halophilic bacteria L-asparaginase. Biologia (Bratisl) 2020. [DOI: 10.2478/s11756-019-00324-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
17
|
Structural basis of the correct subunit assembly, aggregation, and intracellular degradation of nylon hydrolase. Sci Rep 2018; 8:9725. [PMID: 29950566 PMCID: PMC6021441 DOI: 10.1038/s41598-018-27860-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 06/12/2018] [Indexed: 11/09/2022] Open
Abstract
Nylon hydrolase (NylC) is initially expressed as an inactive precursor (36 kDa). The precursor is cleaved autocatalytically at Asn266/Thr267 to generate an active enzyme composed of an α subunit (27 kDa) and a β subunit (9 kDa). Four αβ heterodimers (molecules A-D) form a doughnut-shaped quaternary structure. In this study, the thermostability of the parental NylC was altered by amino acid substitutions located at the A/D interface (D122G/H130Y/D36A/L137A) or the A/B interface (E263Q) and spanned a range of 47 °C. Considering structural, biophysical, and biochemical analyses, we discuss the structural basis of the stability of nylon hydrolase. From the analytical centrifugation data obtained regarding the various mutant enzymes, we conclude that the assembly of the monomeric units is dynamically altered by the mutations. Finally, we propose a model that can predict whether the fate of the nascent polypeptide will be correct subunit assembly, inappropriate protein-protein interactions causing aggregation, or intracellular degradation of the polypeptide.
Collapse
|
18
|
Ajewole E, Santamaria‐Kisiel L, Pajak A, Jaskolski M, Marsolais F. Structural basis of potassium activation in plant asparaginases. FEBS J 2018; 285:1528-1539. [DOI: 10.1111/febs.14428] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 02/07/2018] [Accepted: 02/27/2018] [Indexed: 11/30/2022]
Affiliation(s)
- Ebenezer Ajewole
- Department of Biology University of Western Ontario London Canada
- London Research and Development Centre Agriculture and Agri‐Food Canada London Canada
| | | | - Agnieszka Pajak
- London Research and Development Centre Agriculture and Agri‐Food Canada London Canada
| | - Mariusz Jaskolski
- Center for Biocrystallographic Research Institute of Bioorganic Chemistry Polish Academy of Sciences Poznan Poland
- Department of Crystallography Faculty of Chemistry A. Mickiewicz University Poznan Poland
| | - Frédéric Marsolais
- Department of Biology University of Western Ontario London Canada
- London Research and Development Centre Agriculture and Agri‐Food Canada London Canada
| |
Collapse
|
19
|
Izadpanah Qeshmi F, Homaei A, Fernandes P, Javadpour S. Marine microbial L-asparaginase: Biochemistry, molecular approaches and applications in tumor therapy and in food industry. Microbiol Res 2018; 208:99-112. [PMID: 29551216 DOI: 10.1016/j.micres.2018.01.011] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 01/23/2018] [Accepted: 01/28/2018] [Indexed: 10/18/2022]
Abstract
The marine environment is a rich source of biological and chemical diversity. It covers more than 70% of the Earth's surface and features a wide diversity of habitats, often displaying extreme conditions, where marine organisms thrive, offering a vast pool for microorganisms and enzymes. Given the dissimilarity between marine and terrestrial habitats, enzymes and microorganisms, either novel or with different and appealing features as compared to terrestrial counterparts, may be identified and isolated. L-asparaginase (E.C. 3.5.1.1), is among the relevant enzymes that can be obtained from marine sources. This amidohydrolase acts on L-asparagine and produce L-aspartate and ammonia, accordingly it has an acknowledged chemotherapeutic application, namely in acute lymphoblastic leukemia. Moreover, L-asparaginase is also of interest in the food industry as it prevents acrylamide formation. Terrestrial organisms have been largely tapped for L-asparaginases, but most failed to comply with criteria for practical applications, whereas marine sources have only been marginally screened. This work provides an overview on the relevant features of this enzyme and the framework for its application, with a clear emphasis on the use of L-asparaginase from marine sources. The review envisages to highlight the unique properties of marine L-asparaginases that could make them good candidates for medical applications and industries, especially in food safety.
Collapse
Affiliation(s)
| | - Ahmad Homaei
- Department of Biology, Faculty of Sciences, University of Hormozgan, Bandar Abbas, Iran.
| | - Pedro Fernandes
- Department of Bioengineering and IBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; Faculty of Engineering, Universidade Lusófona de Humanidades e Tecnologias, Av. Campo Grande 376, 1749-024 Lisboa, Portugal
| | - Sedigheh Javadpour
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| |
Collapse
|
20
|
Saeed H, Ali H, Soudan H, Embaby A, El-Sharkawy A, Farag A, Hussein A, Ataya F. Molecular cloning, structural modeling and production of recombinant Aspergillus terreusl. asparaginase in Escherichia coli. Int J Biol Macromol 2018; 106:1041-1051. [PMID: 28851634 DOI: 10.1016/j.ijbiomac.2017.08.110] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 08/16/2017] [Accepted: 08/17/2017] [Indexed: 11/15/2022]
Abstract
l-Asparaginase (EC 3.5.1.1) is an important medical enzyme that catalysis the hydrolysis of l-asparagine to aspartic acid and ammonium. For over four decades l. asparaginase utic agent for the treatment of a variety of lymphoproliferative disorders and lymphoma such as acute lymphoblastic leukemia. In the present study A. terreus full length l. asparaginase gene, 1179bp was optimized for expression in Escherichia coli BL21 (DE3) pLysS. The full length A. terreusl. asparaginase gene encoding a protein of 376 amino acids with estimated molecular weight of 42.0kDa and a theoretical isoelectric point (pI) of 5.0. BLAST and phylogeny analysis revealed that the A. terreusl. asparaginase shared high similarity with other known fungal l. asparaginase (75% homology with A. nomius and 71% with A. nidulans). The recombinant protein was overexpressed in the form of amorphous submicron proteinaceous inclusion bodies upon induction with 1mM IPTG at 37°C for 18h.
Collapse
Affiliation(s)
- Hesham Saeed
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt.
| | - Hadeer Ali
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Hadeer Soudan
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Amira Embaby
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Amany El-Sharkawy
- Marine Biotechnology and Natural Products Extract Laboratory, National Institute of Oceanography and Fisheries (NIOF), Alexandria, Egypt
| | - Aida Farag
- Marine Biotechnology and Natural Products Extract Laboratory, National Institute of Oceanography and Fisheries (NIOF), Alexandria, Egypt
| | - Ahmed Hussein
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Farid Ataya
- Biochemistry Department, College of Science, King Saud University, Bld. 5, Lab AA10, P.O. Box: 2454, Riyadh, Saudi Arabia; Molecular Biology Department, Genetic Engineering Division, National Research Centre, 33 El-Bohouth St. (former El-Tahrir St.), P.O. 12622, Dokki, Giza, Egypt(1)
| |
Collapse
|
21
|
Sushma C, Anand AP, Veeranki VD. Enhanced production of glutaminase free L-asparaginase II by Bacillus subtilis WB800N through media optimization. KOREAN J CHEM ENG 2017. [DOI: 10.1007/s11814-017-0211-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
22
|
Van Kerckhoven SH, de la Torre FN, Cañas RA, Avila C, Cantón FR, Cánovas FM. Characterization of Three L-Asparaginases from Maritime Pine ( Pinus pinaster Ait.). FRONTIERS IN PLANT SCIENCE 2017; 8:1075. [PMID: 28690619 PMCID: PMC5481357 DOI: 10.3389/fpls.2017.01075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 06/06/2017] [Indexed: 05/15/2023]
Abstract
Asparaginases (ASPG, EC 3.5.1.1) catalyze the hydrolysis of the amide group of L-asparagine producing L-aspartate and ammonium. Three ASPG, PpASPG1, PpASPG2, and PpASPG3, have been identified in the transcriptome of maritime pine (Pinus pinaster Ait.) that were transiently expressed in Nicotiana benthamiana by agroinfection. The three recombinant proteins were processed in planta to active enzymes and it was found that all mature forms exhibited double activity asparaginase/isoaspartyl dipeptidase but only PpASPG1 was able to catalyze efficiently L-asparagine hydrolysis. PpASPG1 contains a variable region of 77 amino acids that is critical for proteolytic processing of the precursor and is retained in the mature enzyme. Furthermore, the functional analysis of deletion mutants demonstrated that this protein fragment is required for specific recognition of the substrate and favors enzyme stability. Potassium has a limited effect on the activation of maritime pine ASPG what is consistent with the lack of a critical residue essential for interaction of cation. Taken together, the results presented here highlight the specific features of ASPG from conifers when compared to the enzymes from angiosperms.
Collapse
|
23
|
Rahimzadeh M, Poodat M, Javadpour S, Qeshmi FI, Shamsipour F. Purification, Characterization and Comparison between Two New L-asparaginases from Bacillus PG03 and Bacillus PG04. Open Biochem J 2016; 10:35-45. [PMID: 27999622 PMCID: PMC5144114 DOI: 10.2174/1874091x01610010035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Revised: 08/25/2016] [Accepted: 09/22/2016] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND L-asparaginase has been used as a chemotherapeutic agent in treatment of lymphoblastic leukemia. In the present investigation, Bacillus sp. PG03 and Bacillus sp. PG04 were studied. METHODS L- asparaginases were produced using different culture media and were purified using ion exchange chromatography. RESULTS Maximum productivity was obtained when asparagine was used as the nitrogen source at pH 7 and 48 h after cultivation. New intracellular L-asparaginases showed an apparent molecular weight of 25 kDa and 30 kDa by SDS-PAGE respectively. These enzymes were active in a wide pH range (3-9) with maximum activity at pH 6 for Bacillus PG03 and pH 7 for Bacillus PG04 L-asparaginase. Bacillus PG03 enzyme was optimally active at 37 ˚C and Bacillus PG04 maximum activity was observed at 40˚C. Kinetic parameters km and Vmax of both enzymes were studied using L-asparagine as the substrate. Thermal inactivation studies of Bacillus PG03 and Bacillus PG04 L-asparaginase exhibited t1/2 of 69.3 min and 34.6 min in 37 ˚C respectively. Also T50 and ∆G of inactivation were measured for both enzymes. CONCLUSION The results revealed that both enzymes had appropriate characteristics and thus could be a potential candidate for medical applications.
Collapse
Affiliation(s)
- Mahsa Rahimzadeh
- Molecular Medicine Research Center, Department of Biochemistry, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran; Food and Cosmetic Health Research Center, Department of Biochemistry, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Manijeh Poodat
- Department of Biochemistry, Faculty of Sciences, Payame Noor University of Mashhad, Mashhad, Iran
| | - Sedigheh Javadpour
- Molecular Medicine Research Center, Department of Microbiology, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Fatemeh Izadpanah Qeshmi
- Food and Cosmetic Health Research Center, Department of Biochemistry, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Fereshteh Shamsipour
- Monoclonal Antibody Research Center, Avicenna Research Institute, (ACECR), Tehran, Iran
| |
Collapse
|
24
|
Gaufichon L, Rothstein SJ, Suzuki A. Asparagine Metabolic Pathways in Arabidopsis. PLANT & CELL PHYSIOLOGY 2016; 57:675-89. [PMID: 26628609 DOI: 10.1093/pcp/pcv184] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 11/18/2015] [Indexed: 05/03/2023]
Abstract
Inorganic nitrogen in the form of ammonium is assimilated into asparagine via multiple steps involving glutamine synthetase (GS), glutamate synthase (GOGAT), aspartate aminotransferase (AspAT) and asparagine synthetase (AS) in Arabidopsis. The asparagine amide group is liberated by the reaction catalyzed by asparaginase (ASPG) and also the amino group of asparagine is released by asparagine aminotransferase (AsnAT) for use in the biosynthesis of amino acids. Asparagine plays a primary role in nitrogen recycling, storage and transport in developing and germinating seeds, as well as in vegetative and senescence organs. A small multigene family encodes isoenzymes of each step of asparagine metabolism in Arabidopsis, except for asparagine aminotransferase encoded by a single gene. The aim of this study is to highlight the structure of the genes and encoded enzyme proteins involved in asparagine metabolic pathways; the regulation and role of different isogenes; and kinetic and physiological properties of encoded enzymes in different tissues and developmental stages.
Collapse
Affiliation(s)
- Laure Gaufichon
- INRA, IJPB, UMR1318, ERL CNRS 3559, Saclay Plant Sciences, RD10, F-78026 Versailles, France
| | - Steven J Rothstein
- University of Guelph, Department of Molecular and Cellular Biology, Guelph, Ontario, Canada N1G 2W1
| | - Akira Suzuki
- INRA, IJPB, UMR1318, ERL CNRS 3559, Saclay Plant Sciences, RD10, F-78026 Versailles, France
| |
Collapse
|
25
|
Unraveling the Activation Mechanism of Taspase1 which Controls the Oncogenic AF4-MLL Fusion Protein. EBioMedicine 2015; 2:386-95. [PMID: 26137584 PMCID: PMC4486195 DOI: 10.1016/j.ebiom.2015.04.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 04/14/2015] [Accepted: 04/14/2015] [Indexed: 12/11/2022] Open
Abstract
We have recently demonstrated that Taspase1-mediated cleavage of the AF4–MLL oncoprotein results in the formation of a stable multiprotein complex which forms the key event for the onset of acute proB leukemia in mice. Therefore, Taspase1 represents a conditional oncoprotein in the context of t(4;11) leukemia. In this report, we used site-directed mutagenesis to unravel the molecular events by which Taspase1 becomes sequentially activated. Monomeric pro-enzymes form dimers which are autocatalytically processed into the enzymatically active form of Taspase1 (αββα). The active enzyme cleaves only very few target proteins, e.g., MLL, MLL4 and TFIIA at their corresponding consensus cleavage sites (CSTasp1) as well as AF4–MLL in the case of leukemogenic translocation. This knowledge was translated into the design of a dominant-negative mutant of Taspase1 (dnTASP1). As expected, simultaneous expression of the leukemogenic AF4–MLL and dnTASP1 causes the disappearance of the leukemogenic oncoprotein, because the uncleaved AF4–MLL protein (328 kDa) is subject to proteasomal degradation, while the cleaved AF4–MLL forms a stable oncogenic multi-protein complex with a very long half-life. Moreover, coexpression of dnTASP1 with a BFP-CSTasp1-GFP FRET biosensor effectively inhibits cleavage. The impact of our findings on future drug development and potential treatment options for t(4;11) leukemia will be discussed. Taspase1 has coevolved with the Trithorax/MLL protein family. Taspase1 hydrolyzes MLL and few other substrate proteins at consensus cleavage sites. Taspase1 is a conditional oncoprotein in of solid and hematological cancers. Taspase1 is required for the processing of the leukemogenic AF4–MLL fusion protein. Inhibition of Taspase1 might have a great therapeutic potential.
Collapse
|
26
|
Betti M, García-Calderón M, Pérez-Delgado CM, Credali A, Pal'ove-Balang P, Estivill G, Repčák M, Vega JM, Galván F, Márquez AJ. Reassimilation of ammonium in Lotus japonicus. JOURNAL OF EXPERIMENTAL BOTANY 2014; 65:5557-66. [PMID: 24948681 DOI: 10.1093/jxb/eru260] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
This review summarizes the most recent results obtained in the analysis of two important metabolic pathways involved in the release of internal sources of ammonium in the model legume Lotus japonicus: photorespiratory metabolism and asparagine breakdown mediated by aparaginase (NSE). The use of photorespiratory mutants deficient in plastidic glutamine synthetase (GS2) enabled us to investigate the transcriptomics and metabolomic changes associated with photorespiratory ammonium accumulation in this plant. The results obtained indicate the existence of a coordinate regulation of genes involved in photorespiratory metabolism. Other types of evidence illustrate the multiple interconnections existing among the photorespiratory pathway and other processes such as intermediate metabolism, nodule function, and secondary metabolism in this plant, all of which are substantially affected in GS2-deficient mutants because of the impairment of the photorespiratory cycle. Finally, the importance of asparagine metabolism in L. japonicus is highlighted because of the fact that asparagine constitutes the vast majority of the reduced nitrogen translocated between different organs of this plant. The different types of NSE enzymes and genes which are present in L. japonicus are described. There is a particular focus on the most abundant K(+)-dependent LjNSE1 isoform and how TILLING mutants were used to demonstrate by reverse genetics the importance of this particular isoform in plant growth and seed production.
Collapse
Affiliation(s)
- Marco Betti
- Departamento de Bioquímica Vegetal y Biología Molecular, Facultad de Química, C/ Profesor García González, 1, 41012-Sevilla, Spain
| | - Margarita García-Calderón
- Departamento de Bioquímica Vegetal y Biología Molecular, Facultad de Química, C/ Profesor García González, 1, 41012-Sevilla, Spain
| | - Carmen M Pérez-Delgado
- Departamento de Bioquímica Vegetal y Biología Molecular, Facultad de Química, C/ Profesor García González, 1, 41012-Sevilla, Spain
| | - Alfredo Credali
- Departamento de Bioquímica Vegetal y Biología Molecular, Facultad de Química, C/ Profesor García González, 1, 41012-Sevilla, Spain
| | - Peter Pal'ove-Balang
- Institute of Biology and Ecology, Faculty of Science, P.J. Šafárik University, Mánesova 23, SK-04001 Košice, Slovak Republic
| | - Guillermo Estivill
- Departamento de Bioquímica Vegetal y Biología Molecular, Facultad de Química, C/ Profesor García González, 1, 41012-Sevilla, Spain
| | - Miroslav Repčák
- Institute of Biology and Ecology, Faculty of Science, P.J. Šafárik University, Mánesova 23, SK-04001 Košice, Slovak Republic
| | - José M Vega
- Departamento de Bioquímica Vegetal y Biología Molecular, Facultad de Química, C/ Profesor García González, 1, 41012-Sevilla, Spain
| | - Francisco Galván
- Departamento de Bioquímica Vegetal y Biología Molecular, Facultad de Química, C/ Profesor García González, 1, 41012-Sevilla, Spain
| | - Antonio J Márquez
- Departamento de Bioquímica Vegetal y Biología Molecular, Facultad de Química, C/ Profesor García González, 1, 41012-Sevilla, Spain
| |
Collapse
|
27
|
Borek D, Kozak M, Pei J, Jaskolski M. Crystal structure of active site mutant of antileukemicl-asparaginase reveals conserved zinc-binding site. FEBS J 2014; 281:4097-111. [DOI: 10.1111/febs.12906] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 06/16/2014] [Accepted: 07/01/2014] [Indexed: 02/03/2023]
Affiliation(s)
- Dominika Borek
- Department of Crystallography; Faculty of Chemistry; A. Mickiewicz University; Poznan Poland
| | - Maciej Kozak
- Department of Crystallography; Faculty of Chemistry; A. Mickiewicz University; Poznan Poland
- Department of Macromolecular Physics; Faculty of Physics; A. Mickiewicz University; Poznan Poland
| | - Jimin Pei
- Howard Hughes Medical Institute; University of Texas Southwestern Medical Center; Dallas TX USA
| | - Mariusz Jaskolski
- Department of Crystallography; Faculty of Chemistry; A. Mickiewicz University; Poznan Poland
- Center for Biocrystallographic Research; Institute of Bioorganic Chemistry; Polish Academy of Sciences; Poznan Poland
| |
Collapse
|
28
|
Bejger M, Imiolczyk B, Clavel D, Gilski M, Pajak A, Marsolais F, Jaskolski M. Na⁺/K⁺ exchange switches the catalytic apparatus of potassium-dependent plant L-asparaginase. ACTA ACUST UNITED AC 2014; 70:1854-72. [PMID: 25004963 DOI: 10.1107/s1399004714008700] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 04/16/2014] [Indexed: 01/03/2023]
Abstract
Plant-type L-asparaginases, which are a subclass of the Ntn-hydrolase family, are divided into potassium-dependent and potassium-independent enzymes with different substrate preferences. While the potassium-independent enzymes have already been well characterized, there are no structural data for any of the members of the potassium-dependent group to illuminate the intriguing dependence of their catalytic mechanism on alkali-metal cations. Here, three crystal structures of a potassium-dependent plant-type L-asparaginase from Phaseolus vulgaris (PvAspG1) differing in the type of associated alkali metal ions (K(+), Na(+) or both) are presented and the structural consequences of the different ions are correlated with the enzyme activity. As in all plant-type L-asparaginases, immature PvAspG1 is a homodimer of two protein chains, which both undergo autocatalytic cleavage to α and β subunits, thus creating the mature heterotetramer or dimer of heterodimers (αβ)2. The αβ subunits of PvAspG1 are folded similarly to the potassium-independent enzymes, with a sandwich of two β-sheets flanked on each side by a layer of helices. In addition to the `sodium loop' (here referred to as the `stabilization loop') known from potassium-independent plant-type asparaginases, the potassium-dependent PvAspG1 enzyme contains another alkali metal-binding loop (the `activation loop') in subunit α (residues Val111-Ser118). The active site of PvAspG1 is located between these two metal-binding loops and in the immediate neighbourhood of three residues, His117, Arg224 and Glu250, acting as a catalytic switch, which is a novel feature that is identified in plant-type L-asparaginases for the first time. A comparison of the three PvAspG1 structures demonstrates how the metal ion bound in the activation loop influences its conformation, setting the catalytic switch to ON (when K(+) is coordinated) or OFF (when Na(+) is coordinated) to respectively allow or prevent anchoring of the reaction substrate/product in the active site. Moreover, it is proposed that Ser118, the last residue of the activation loop, is involved in the potassium-dependence mechanism. The PvAspG1 structures are discussed in comparison with those of potassium-independent L-asparaginases (LlA, EcAIII and hASNase3) and those of other Ntn-hydrolases (AGA and Tas1), as well as in the light of noncrystallographic studies.
Collapse
Affiliation(s)
- Magdalena Bejger
- Center for Biocrystallographic Research, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Barbara Imiolczyk
- Center for Biocrystallographic Research, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Damien Clavel
- Department of Crystallography, Faculty of Chemistry, A. Mickiewicz University, Poznan, Poland
| | - Miroslaw Gilski
- Center for Biocrystallographic Research, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | | | | | - Mariusz Jaskolski
- Center for Biocrystallographic Research, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| |
Collapse
|
29
|
Nomme J, Su Y, Lavie A. Elucidation of the specific function of the conserved threonine triad responsible for human L-asparaginase autocleavage and substrate hydrolysis. J Mol Biol 2014; 426:2471-85. [PMID: 24768817 DOI: 10.1016/j.jmb.2014.04.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 04/14/2014] [Accepted: 04/15/2014] [Indexed: 02/05/2023]
Abstract
Our long-term goal is the design of a human l-asparaginase (hASNase3) variant, suitable for use in cancer therapy without the immunogenicity problems associated with the currently used bacterial enzymes. Asparaginases catalyze the hydrolysis of the amino acid asparagine to aspartate and ammonia. The key property allowing for the depletion of blood asparagine by bacterial asparaginases is their low micromolar KM value. In contrast, human enzymes have a millimolar KM for asparagine. Toward the goal of engineering an hASNase3 variant with micromolar KM, we conducted a structure/function analysis of the conserved catalytic threonine triad of this human enzyme. As a member of the N-terminal nucleophile family, to become enzymatically active, hASNase3 must undergo autocleavage between residues Gly167 and Thr168. To determine the individual contribution of each of the three conserved active-site threonines (threonine triad Thr168, Thr186, Thr219) for the enzyme-activating autocleavage and asparaginase reactions, we prepared the T168S, T186V and T219A/V mutants. These mutants were tested for their ability to cleave and to catalyze asparagine hydrolysis, in addition to being examined structurally. We also elucidated the first N-terminal nucleophile plant-type asparaginase structure in the covalent intermediate state. Our studies indicate that, while not all triad threonines are required for the cleavage reaction, all are essential for the asparaginase activity. The increased understanding of hASNase3 function resulting from these studies reveals the key regions that govern cleavage and the asparaginase reaction, which may inform the design of variants that attain a low KM for asparagine.
Collapse
Affiliation(s)
- Julian Nomme
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Ying Su
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Arnon Lavie
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA.
| |
Collapse
|
30
|
Schalk AM, Lavie A. Structural and kinetic characterization of guinea pig L-asparaginase type III. Biochemistry 2014; 53:2318-28. [PMID: 24669941 PMCID: PMC4004260 DOI: 10.1021/bi401692v] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We investigated whether an uncharacterized protein from guinea pig could be the enzyme behind Kidd's serendipitous discovery, made over 60 years ago, that guinea pig serum has cell killing ability. It has been long known that an enzyme with l-asparaginase activity is responsible for cell killing, although astonishingly, its identity remains unclear. Bacterial asparaginases with similar cell killing properties have since become a mainstay therapy of certain cancers such as acute lymphoblastic leukemia. By hydrolyzing asparagine to aspartate and ammonia, these drugs deplete the asparagine present in the blood, killing cancer cells that rely on extracellular asparagine uptake for survival. However, bacterial asparaginases can elicit an adverse immune response. We propose that replacement of bacterial enzymes with the guinea pig asparaginase responsible for serum activity, by its virtue of being more closely related to human enzymes, will be less immunogenic. To this goal, we investigated whether an uncharacterized protein from guinea pig with putative asparaginase activity, which we call gpASNase3, could be that enzyme. We examined its self-activation process (gpASNase3 requires autocleavage to become active), kinetically characterized it for asparaginase and β-aspartyl dipeptidase activity, and elucidated its crystal structure in both the uncleaved and cleaved states. This work reveals that gpASNase3 is not the enzyme responsible for the antitumor effects of guinea pig serum. It exhibits a low affinity for asparagine as measured by a high Michaelis constant, KM, in the millimolar range, in contrast to the low KM (micromolar range) required for asparaginase to be effective as an anticancer agent.
Collapse
Affiliation(s)
- Amanda M Schalk
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago , 900 S. Ashland, Chicago , Illinois 60607, United States
| | | |
Collapse
|
31
|
Su Y, Karamitros CS, Nomme J, McSorley T, Konrad M, Lavie A. Free glycine accelerates the autoproteolytic activation of human asparaginase. ACTA ACUST UNITED AC 2013; 20:533-40. [PMID: 23601642 DOI: 10.1016/j.chembiol.2013.03.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 02/25/2013] [Accepted: 03/04/2013] [Indexed: 12/21/2022]
Abstract
Human asparaginase 3 (hASNase3), which belongs to the N-terminal nucleophile hydrolase superfamily, is synthesized as a single polypeptide that is devoid of asparaginase activity. Intramolecular autoproteolytic processing releases the amino group of Thr168, a moiety required for catalyzing asparagine hydrolysis. Recombinant hASNase3 purifies as the uncleaved, asparaginase-inactive form and undergoes self-cleavage to the active form at a very slow rate. Here, we show that the free amino acid glycine selectively acts to accelerate hASNase3 cleavage both in vitro and in human cells. Other small amino acids such as alanine, serine, or the substrate asparagine are not capable of promoting autoproteolysis. Crystal structures of hASNase3 in complex with glycine in the uncleaved and cleaved enzyme states reveal the mechanism of glycine-accelerated posttranslational processing and explain why no other amino acid can substitute for glycine.
Collapse
Affiliation(s)
- Ying Su
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | | | | | | | | | | |
Collapse
|
32
|
Li W, Cantor JR, Yogesha S, Yang S, Chantranupong L, Liu JQ, Agnello G, Georgiou G, Stone EM, Zhang Y. Uncoupling intramolecular processing and substrate hydrolysis in the N-terminal nucleophile hydrolase hASRGL1 by circular permutation. ACS Chem Biol 2012; 7:1840-7. [PMID: 22891768 PMCID: PMC3514461 DOI: 10.1021/cb300232n] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The human asparaginase-like protein 1 (hASRGL1) catalyzes the hydrolysis of l-asparagine and isoaspartyl-dipeptides. As an N-terminal nucleophile (Ntn) hydrolase superfamily member, the active form of hASRGL1 is generated by an intramolecular cleavage step with Thr168 as the catalytic residue. However, in vitro, autoprocessing is incomplete (~50%), fettering the biophysical characterization of hASRGL1. We circumvented this obstacle by constructing a circularly permuted hASRGL1 that uncoupled the autoprocessing reaction, allowing us to kinetically and structurally characterize this enzyme and the precursor-like hASRGL1-Thr168Ala variant. Crystallographic and biochemical evidence suggest an activation mechanism where a torsional restraint on the Thr168 side chain helps drive the intramolecular processing reaction. Cleavage and formation of the active site releases the torsional restriction on Thr168, which is facilitated by a small conserved Gly-rich loop near the active site that allows the conformational changes necessary for activation.
Collapse
Affiliation(s)
- Wenzong Li
- Department of Chemistry and Biochemistry, University of Texas, Austin, Texas 78712
| | - Jason R Cantor
- Departments of Biomedical and Chemical Engineering, University of Texas, Austin, Texas 78712
| | - S.D. Yogesha
- Department of Chemistry and Biochemistry, University of Texas, Austin, Texas 78712
| | - Shirley Yang
- Departments of Biomedical and Chemical Engineering, University of Texas, Austin, Texas 78712
| | - Lynne Chantranupong
- Departments of Biomedical and Chemical Engineering, University of Texas, Austin, Texas 78712
| | - June Qingxia Liu
- Department of Chemistry and Biochemistry, University of Texas, Austin, Texas 78712
| | - Giulia Agnello
- Institute of Cellular and Molecular Biology, University of Texas, Austin, Texas 78712
| | - George Georgiou
- Departments of Biomedical and Chemical Engineering, University of Texas, Austin, Texas 78712
,Section of Molecular Genetics and Microbiology, University of Texas, Austin, Texas 78712
,Institute of Cellular and Molecular Biology, University of Texas, Austin, Texas 78712
| | - Everett M Stone
- Departments of Biomedical and Chemical Engineering, University of Texas, Austin, Texas 78712
,Address correspondence to: Yan Zhang: 1 University Station A5300, Austin, TX 78712. Phone: (512)-471-8645. Fax: 512-471-9469. or Everett Stone: 1 University Station C0800, Austin, TX 78712. Phone: (512) 512-232-4105. stonesci@.utexas.edu
| | - Yan Zhang
- Department of Chemistry and Biochemistry, University of Texas, Austin, Texas 78712
,Institute of Cellular and Molecular Biology, University of Texas, Austin, Texas 78712
,Address correspondence to: Yan Zhang: 1 University Station A5300, Austin, TX 78712. Phone: (512)-471-8645. Fax: 512-471-9469. or Everett Stone: 1 University Station C0800, Austin, TX 78712. Phone: (512) 512-232-4105. stonesci@.utexas.edu
| |
Collapse
|
33
|
Nomme J, Su Y, Konrad M, Lavie A. Structures of apo and product-bound human L-asparaginase: insights into the mechanism of autoproteolysis and substrate hydrolysis. Biochemistry 2012; 51:6816-26. [PMID: 22861376 DOI: 10.1021/bi300870g] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Asparaginases catalyze the hydrolysis of the amino acid asparagine to aspartate and ammonia. Bacterial asparaginases are used in cancer chemotherapy to deplete asparagine from the blood, because several hematological malignancies depend on extracellular asparagine for growth. To avoid the immune response against the bacterial enzymes, it would be beneficial to replace them with human asparaginases. However, unlike the bacterial asparaginases, the human enzymes have a millimolar K(m) value for asparagine, making them inefficient in depleting the amino acid from blood. To facilitate the development of human variants suitable for therapeutic use, we determined the structure of human l-asparaginase (hASNase3). This asparaginase is an N-terminal nucleophile (Ntn) family member that requires autocleavage between Gly167 and Thr168 to become catalytically competent. For most Ntn hydrolases, this autoproteolytic activation occurs efficiently. In contrast, hASNas3 is relatively stable in its uncleaved state, and this allowed us to observe the structure of the enzyme prior to cleavage. To determine the structure of the cleaved state, we exploited our discovery that the free amino acid glycine promotes complete cleavage of hASNase3. Both enzyme states were elucidated in the absence and presence of the product aspartate. Together, these structures provide insight into the conformational changes required for cleavage and the precise enzyme-substrate interactions. The new understanding of hASNase3 will serve to guide the design of variants that possess a decreased K(m) value for asparagine, making the human enzyme a suitable replacement for the bacterial asparaginases in cancer therapy.
Collapse
Affiliation(s)
- Julian Nomme
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | | | | | | |
Collapse
|
34
|
Gabriel M, Telmer PG, Marsolais F. Role of asparaginase variable loop at the carboxyl terminal of the alpha subunit in the determination of substrate preference in plants. PLANTA 2012; 235:1013-1022. [PMID: 22127737 DOI: 10.1007/s00425-011-1557-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Accepted: 11/17/2011] [Indexed: 05/27/2023]
Abstract
Structural determinants responsible for the substrate preference of the potassium-independent (ASPGA1) and -dependent (ASPGB1) asparaginases from Arabidopsis thaliana have been investigated. Like ASPGA1, ASPGB1 was found to be catalytically active with both L: -Asn and β-Asp-His as substrates, contrary to a previous report. However, ASPGB1 had a 45-fold higher specific activity with Asn as substrate than ASPGA1. A divergent sequence between the two enzymes forms a variable loop at the C-terminal of the alpha subunit. The results of dynamic simulations have previously implicated a movement of the C-terminus in the allosteric transduction of K(+)-binding at the surface of LjNSE1 asparaginase. In the crystal structure of Lupinus luteus asparaginase, most residues in this segment cannot be visualized due to a weak electron density. Exchanging the variable loop in ASPGA1 with that from ASPGB1 increased the affinity for Asn, with a 320-fold reduction in K (m) value. Homology modeling identified a residue specific to ASPGB1, Phe(162), preceding the variable loop, whose side chain is located in proximity to the beta-carboxylate group of the product aspartate, and to Gly(246), a residue participating in an oxyanion hole which stabilizes a negative charge forming on the side chain oxygen of asparagine during catalysis. Replacement with the corresponding leucine from ASPGA1 specifically lowered the V (max) value with Asn as substrate by 8.4-fold.
Collapse
Affiliation(s)
- Michelle Gabriel
- Department of Biology, University of Western Ontario, London, ON, Canada
| | | | | |
Collapse
|
35
|
Oza VP, Parmar PP, Patel DH, Subramanian RB. Cloning, expression and characterization of l-asparaginase from Withania somnifera L. for large scale production. 3 Biotech 2011; 1:21-26. [PMID: 22558532 PMCID: PMC3339582 DOI: 10.1007/s13205-011-0003-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2010] [Accepted: 02/16/2011] [Indexed: 11/30/2022] Open
Abstract
l-Asparaginase (E.C. 3.5.1.1) is used as a therapeutic agent in the treatment of acute childhood lymphoblastic leukemia. It is found in a variety of organisms such as microbes, plants and mammals. In plants, l-asparaginase enzymes are required to catalyze the release of ammonia from asparagine, which is the main nitrogen-relocation molecule in these organisms. An Indian medicinal plant, Withania somnifera was reported as a novel source of l-asparaginase. l-Asparaginase from W. somnifera was cloned and overexpressed in E. coli. The enzymatic properties of the recombinant enzyme were investigated and the kinetic parameters (K(m), k(cat)) for a number of substrates were determined. The kinetic parameters of selected substrates were determined at various pH and the pH- and temperature-dependence profiles were analyzed. WA gene successfully cloned into E. coli BL21 (DE3) showed high asparaginase activity with a specific activity of 17.3 IU/mg protein.
Collapse
Affiliation(s)
- Vishal P. Oza
- Department of Plant Biotechnology, B R D School of Biosciences, Sardar Patel Maidan, Vadtal Road, Satellite Campus, Sardar Patel University, Post Box No. 39, Vallabh Vidyanagar, 388 120 Gujarat India
| | - Pritesh P. Parmar
- Department of Plant Biotechnology, B R D School of Biosciences, Sardar Patel Maidan, Vadtal Road, Satellite Campus, Sardar Patel University, Post Box No. 39, Vallabh Vidyanagar, 388 120 Gujarat India
| | - Darshan H. Patel
- Department of Plant Biotechnology, B R D School of Biosciences, Sardar Patel Maidan, Vadtal Road, Satellite Campus, Sardar Patel University, Post Box No. 39, Vallabh Vidyanagar, 388 120 Gujarat India
| | - R. B. Subramanian
- Department of Plant Biotechnology, B R D School of Biosciences, Sardar Patel Maidan, Vadtal Road, Satellite Campus, Sardar Patel University, Post Box No. 39, Vallabh Vidyanagar, 388 120 Gujarat India
| |
Collapse
|
36
|
Credali A, Díaz-Quintana A, García-Calderón M, De la Rosa MA, Márquez AJ, Vega JM. Structural analysis of K+ dependence in L-asparaginases from Lotus japonicus. PLANTA 2011; 234:109-22. [PMID: 21390508 DOI: 10.1007/s00425-011-1393-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Accepted: 02/23/2011] [Indexed: 05/11/2023]
Abstract
The molecular features responsible for the existence in plants of K+-dependent asparaginases have been investigated. For this purpose, two different cDNAs were isolated in Lotus japonicus, encoding for K+-dependent (LjNSE1) or K+-independent (LjNSE2) asparaginases. Recombinant proteins encoded by these cDNAs have been purified and characterized. Both types of asparaginases are composed by two different subunits, α (20 kDa) and β (17 kDa), disposed as (αβ)₂ quaternary structure. Major differences were found in the catalytic efficiency of both enzymes, due to the fact that K+ is able to increase by tenfold the enzyme activity and lowers the K(m) for asparagine specifically in LjNSE1 but not in LjNSE2 isoform. Optimum LjNSE1 activity was found at 5-50 mM K+, with a K(m) for K+ of 0.25 mM. Na+ and Rb+ can, to some extent, substitute for K+ on the activating effect of LjNSE1 more efficiently than Cs+ and Li+ does. In addition, K+ is able to stabilize LjNSE1 against thermal inactivation. Protein homology modelling and molecular dynamics studies, complemented with site-directed mutagenesis, revealed the key importance of E248, D285 and E286 residues for the catalytic activity and K+ dependence of LjNSE1, as well as the crucial relevance of K+ for the proper orientation of asparagine substrate within the enzyme molecule. On the other hand, LjNSE2 but not LjNSE1 showed β-aspartyl-hydrolase activity (K(m) = 0.54 mM for β-Asp-His). These results are discussed in terms of the different physiological significance of these isoenzymes in plants.
Collapse
Affiliation(s)
- Alfredo Credali
- Departamento de Bioquímica Vegetal y Biología Molecular, Facultad de Química, Universidad de Sevilla, 41071 Seville, Spain
| | | | | | | | | | | |
Collapse
|
37
|
Yin J, Deng Z, Zhao G, Huang X. The N-terminal nucleophile serine of cephalosporin acylase executes the second autoproteolytic cleavage and acylpeptide hydrolysis. J Biol Chem 2011; 286:24476-86. [PMID: 21576250 DOI: 10.1074/jbc.m111.242313] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cephalosporin acylase (CA) precursor is translated as a single polypeptide chain and folds into a self-activating pre-protein. Activation requires two peptide bond cleavages that excise an internal spacer to form the mature αβ heterodimer. Using Q-TOF LC-MS, we located the second cleavage site between Glu(159) and Gly(160), and detected the corresponding 10-aa spacer (160)GDPPDLADQG(169) of CA mutants. The site of the second cleavage depended on Glu(159): moving Glu into the spacer or removing 5-10 residues from the spacer sequence resulted in shorter spacers with the cleavage at the carboxylic side of Glu. The mutant E159D was cleaved more slowly than the wild-type, as were mutants G160A and G160L. This allowed kinetic measurements showing that the second cleavage reaction was a first-order, intra-molecular process. Glutaryl-7-aminocephalosporanic acid is the classic substrate of CA, in which the N-terminal Ser(170) of the β-subunit, is the nucleophile. Glu and Asp resemble glutaryl, suggesting that CA might also remove N-terminal Glu or Asp from peptides. This was indeed the case, suggesting that the N-terminal nucleophile also performed the second proteolytic cleavage. We also found that CA is an acylpeptide hydrolase rather than a previously expected acylamino acid acylase. It only exhibited exopeptidase activity for the hydrolysis of an externally added peptide, supporting the intra-molecular interaction. We propose that the final CA activation is an intra-molecular process performed by an N-terminal nucleophile, during which large conformational changes in the α-subunit C-terminal region are required to bridge the gap between Glu(159) and Ser(170).
Collapse
Affiliation(s)
- Jun Yin
- State Key Laboratory of Microbial Metabolism, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai 200030, China
| | | | | | | |
Collapse
|
38
|
Lakomek K, Dickmanns A, Kettwig M, Urlaub H, Ficner R, Lübke T. Initial insight into the function of the lysosomal 66.3 kDa protein from mouse by means of X-ray crystallography. BMC STRUCTURAL BIOLOGY 2009; 9:56. [PMID: 19706171 PMCID: PMC2739207 DOI: 10.1186/1472-6807-9-56] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Accepted: 08/25/2009] [Indexed: 11/24/2024]
Abstract
Background The lysosomal 66.3 kDa protein from mouse is a soluble, mannose 6-phosphate containing protein of so far unknown function. It is synthesized as a glycosylated 75 kDa precursor that undergoes limited proteolysis leading to a 28 kDa N- and a 40 kDa C-terminal fragment. Results In order to gain insight into the function and the post-translational maturation process of the glycosylated 66.3 kDa protein, three crystal structures were determined that represent different maturation states. These structures demonstrate that the 28 kDa and 40 kDa fragment which have been derived by a proteolytic cleavage remain associated. Mass spectrometric analysis confirmed the subsequent trimming of the C-terminus of the 28 kDa fragment making a large pocket accessible, at the bottom of which the putative active site is located. The crystal structures reveal a significant similarity of the 66.3 kDa protein to several bacterial hydrolases. The core αββα sandwich fold and a cysteine residue at the N-terminus of the 40 kDa fragment (C249) classify the 66.3 kDa protein as a member of the structurally defined N-terminal nucleophile (Ntn) hydrolase superfamily. Conclusion Due to the close resemblance of the 66.3 kDa protein to members of the Ntn hydrolase superfamily a hydrolytic activity on substrates containing a non-peptide amide bond seems reasonable. The structural homology which comprises both the overall fold and essential active site residues also implies an autocatalytic maturation process of the lysosomal 66.3 kDa protein. Upon the proteolytic cleavage between S248 and C249, a deep pocket becomes solvent accessible, which harbors the putative active site of the 66.3 kDa protein.
Collapse
Affiliation(s)
- Kristina Lakomek
- Department of Molecular Structural Biology, Institute of Microbiology and Genetics, GZMB, Georg-August University Goettingen, Justus-von-Liebig-Weg 11, D-37077 Goettingen, Germany.
| | | | | | | | | | | |
Collapse
|
39
|
Iqbal A, Clifton IJ, Bagonis M, Kershaw NJ, Domene C, Claridge TDW, Wharton CW, Schofield CJ. Anatomy of a Simple Acyl Intermediate in Enzyme Catalysis: Combined Biophysical and Modeling Studies on Ornithine Acetyl Transferase. J Am Chem Soc 2008; 131:749-57. [DOI: 10.1021/ja807215u] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Aman Iqbal
- Chemistry Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, U.K., Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, OX1 3QZ, U.K., School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, U.K
| | - Ian J. Clifton
- Chemistry Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, U.K., Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, OX1 3QZ, U.K., School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, U.K
| | - Maria Bagonis
- Chemistry Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, U.K., Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, OX1 3QZ, U.K., School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, U.K
| | - Nadia J. Kershaw
- Chemistry Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, U.K., Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, OX1 3QZ, U.K., School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, U.K
| | - Carmen Domene
- Chemistry Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, U.K., Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, OX1 3QZ, U.K., School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, U.K
| | - Timothy D. W. Claridge
- Chemistry Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, U.K., Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, OX1 3QZ, U.K., School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, U.K
| | - Christopher W. Wharton
- Chemistry Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, U.K., Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, OX1 3QZ, U.K., School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, U.K
| | - Christopher J. Schofield
- Chemistry Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, U.K., Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, OX1 3QZ, U.K., School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, U.K
| |
Collapse
|
40
|
Abstract
This article comprises detailed information about L-asparaginase, encompassing topics such as microbial and plant sources of L-asparaginase, treatment with L-asparaginase, mechanism of action of L-asparaginase, production, purification, properties, expression and characteristics of l-asparaginase along with information about studies on the structure of L-asparaginase. Although L-asparaginase has been reviewed by Savitri and Azmi (2003), our effort has been to include recent and updated information about the enzyme covering new aspects such as structural modification and immobilization of L-asparaginase, recombinant L-asparaginase, resistance to L-asparaginase, methods of assay of L-asparagine and L-asparaginase activity using the biosensor approach, L-asparaginase activity in soil and the factors affecting it. Also, side-effects of L-asparaginase treatment in acute lymphoblastic leukemia (ALL) have been discussed in the current review. L-asparaginase has been and is still one of the most widely studied therapeutic enzymes by researchers and scientists worldwide.
Collapse
Affiliation(s)
- Neelam Verma
- Biosensor Technology Lab, Department of Biotechnology, Punjabi University, Patiala, Punjab, India.
| | | | | | | |
Collapse
|
41
|
Michalska K, Hernandez-Santoyo A, Jaskolski M. The Mechanism of Autocatalytic Activation of Plant-type L-Asparaginases. J Biol Chem 2008; 283:13388-97. [DOI: 10.1074/jbc.m800746200] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
42
|
Gherardini PF, Wass MN, Helmer-Citterich M, Sternberg MJE. Convergent Evolution of Enzyme Active Sites Is not a Rare Phenomenon. J Mol Biol 2007; 372:817-45. [PMID: 17681532 DOI: 10.1016/j.jmb.2007.06.017] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2006] [Revised: 05/14/2007] [Accepted: 06/08/2007] [Indexed: 02/03/2023]
Abstract
Since convergent evolution of enzyme active sites was first identified in serine proteases, other individual instances of this phenomenon have been documented. However, a systematic analysis assessing the frequency of this phenomenon across enzyme space is still lacking. This work uses the Query3d structural comparison algorithm to integrate for the first time detailed knowledge about catalytic residues, available through the Catalytic Site Atlas (CSA), with the evolutionary information provided by the Structural Classification of Proteins (SCOP) database. This study considers two modes of convergent evolution: (i) mechanistic analogues which are enzymes that use the same mechanism to perform related, but possibly different, reactions (considered here as sharing the first three digits of the EC number); and (ii) transformational analogues which catalyse exactly the same reaction (identical EC numbers), but may use different mechanisms. Mechanistic analogues were identified in 15% (26 out of 169) of the three-digit EC groups considered, showing that this phenomenon is not rare. Furthermore 11 of these groups also contain transformational analogues. The catalytic triad is the most widespread active site; the results of the structural comparison show that this mechanism, or variations thereof, is present in 23 superfamilies. Transformational analogues were identified for 45 of the 951 four-digit EC numbers present within the CSA and about half of these were also mechanistic analogues exhibiting convergence of their active sites. This analysis has also been extended to the whole Protein Data Bank to provide a complete and manually curated list of the all the transformational analogues whose structure is classified in SCOP. The results of this work show that the phenomenon of convergent evolution is not rare, especially when considering large enzymatic families.
Collapse
Affiliation(s)
- Pier Federico Gherardini
- Biochemistry Building, Division of Molecular Biosciences, Imperial College London, London SW7 2AZ, UK
| | | | | | | |
Collapse
|
43
|
Davies JQ, Chang GW, Yona S, Gordon S, Stacey M, Lin HH. The Role of Receptor Oligomerization in Modulating the Expression and Function of Leukocyte Adhesion-G Protein-coupled Receptors. J Biol Chem 2007; 282:27343-27353. [PMID: 17620333 DOI: 10.1074/jbc.m704096200] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The human leukocyte adhesion-G protein-coupled receptors (GPCRs), the epidermal growth factor (EGF)-TM7 proteins, are shown here to function as homo- and hetero-oligomers. Using cell surface cross-linking, co-immunoprecipitation, and fluorescence resonance energy transfer analysis of EMR2, an EGF-TM7 receptor predominantly expressed in myeloid cells, we demonstrate that it forms dimers in a reaction mediated exclusively by the TM7 moiety. We have also identified a naturally occurring but structurally unstable EMR2 splice variant that acts as a dominant negative modulator by dimerizing with the wild type receptor and down-regulating its expression. Additionally, heterodimerization between closely related EGF-TM7 members is shown to result in the modulation of expression and ligand binding properties of the receptors. These findings suggest that receptor homo- and hetero-oligomerization play a regulatory role in modulating the expression and function of leukocyte adhesion-GPCRs.
Collapse
Affiliation(s)
- John Q Davies
- Sir William Dunn School of Pathology, University of Oxford, South Parks Rd., Oxford, OX1 3RE, United Kingdom and the
| | - Gin-Wen Chang
- Sir William Dunn School of Pathology, University of Oxford, South Parks Rd., Oxford, OX1 3RE, United Kingdom and the
| | - Simon Yona
- Sir William Dunn School of Pathology, University of Oxford, South Parks Rd., Oxford, OX1 3RE, United Kingdom and the
| | - Siamon Gordon
- Sir William Dunn School of Pathology, University of Oxford, South Parks Rd., Oxford, OX1 3RE, United Kingdom and the
| | - Martin Stacey
- Sir William Dunn School of Pathology, University of Oxford, South Parks Rd., Oxford, OX1 3RE, United Kingdom and the.
| | - Hsi-Hsien Lin
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Rd., Kwei-San, Tao-Yuan, Taiwan.
| |
Collapse
|
44
|
O'Rourke JA, Graham MA, Vodkin L, Gonzalez DO, Cianzio SR, Shoemaker RC. Recovering from iron deficiency chlorosis in near-isogenic soybeans: a microarray study. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2007; 45:287-92. [PMID: 17466527 DOI: 10.1016/j.plaphy.2007.03.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Iron deficiency chlorosis (IDC) in soybeans has proven to be a perennial problem in the calcareous soils of the U.S. upper Midwest. A historically difficult trait to study in fields, the use of hydroponics in a controlled greenhouse environment has provided a mechanism to study genetic variation while limiting environmental complications. IDC susceptible plants growing in calcareous soils and in iron-controlled hydroponic experiments often exhibit a characteristic chlorotic phenotype early in the growing season but are able to re-green later in the season. To examine the changes in gene expression of these plants, near-isogenic lines, iron efficient PI548553 (Clark) and iron inefficient PI547430 (IsoClark), developed for their response to iron deficiency stress [USDA, ARS, National Genetic Resources Program, Germplasm Resources Information Network - GRIN. (Online Database) National Germplasm Resources Laboratory, Beltsville, MD, 2004. Available: http://www.ars.grin.gov/cgi-bin/npgs/html/acc_search.pl?accid=PI+547430. [22] were grown in iron-deficient hydroponic conditions for one week, then transferred to iron sufficient conditions for another week. This induced a phenotypic response mimicking the growth of the plants in the field; initial chlorosis followed by re-greening. RNA was isolated from root tissue and transcript profiles were examined between the two near-isogenic lines using publicly available cDNA microarrays. By alleviating the iron deficiency stress our expectation was that plants would return to baseline expression levels. However, the microarray comparison identified four cDNAs that were under-expressed by a two-fold or greater difference in the iron inefficient plant compared to the iron efficient plant. This differential expression was re-examined and confirmed by real time PCR experimentation. Control experiments showed that these genes are not differentially expressed in plants grown continually under iron rich hydroponic conditions. The expression differences suggest potential residual effects of iron deficiency on plant health.
Collapse
Affiliation(s)
- Jamie A O'Rourke
- Department of Genetics, Developmental and Cellular Biology, Iowa State University, Ames, IA 50011, USA
| | | | | | | | | | | |
Collapse
|
45
|
Cañas RA, de la Torre F, Cánovas FM, Cantón FR. Coordination of PsAS1 and PsASPG expression controls timing of re-allocated N utilization in hypocotyls of pine seedlings. PLANTA 2007; 225:1205-19. [PMID: 17123103 DOI: 10.1007/s00425-006-0431-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2006] [Accepted: 10/13/2006] [Indexed: 05/11/2023]
Abstract
During pine seed germination, a large amount of N mobilized from the storage proteins is re-allocated in the hypocotyl as free asparagine, as a result of the high levels of asparagine synthetase (AS) encoded by the PsAS1 gene. To determine the role of this re-allocated N reserve, a full-length cDNA encoding L: -asparaginase (ASPG) has been cloned from Scots pine (Pinus sylvestris L.) seedlings and characterized. Like other N-terminal nucleophile hydrolases, pine ASPG requires a post-translational processing to exhibit enzymatic activity. However, in contrast to previous reports on other plant ASPGs, purified recombinant pine ASPG does not undergo autoproteolytic cleavage in vitro. Our results suggest that the processing requires accessory proteins to assist in the proteolysis or in the proper folding before autocleavage in a divalent cation-dependent manner. Sequence comparison analysis revealed that the pine protein is included in the K+-dependent subfamily of plant ASPGs. The expression of the ASPG-encoding gene (PsASPG) was higher in organs with extensive secondary development of the vascular system. The increase in transcript abundance observed at advanced stages of hypocotyl development was concomitant with a decrease of PsAS1 transcript abundance and a remarkable increase in the number of xylem elements and highly lignified cell walls. These results, together with the precise localization of PsASPG transcripts in cells of the cambial region, suggest that the expression of PsAS1 and PsASPG is temporally coordinated, to control the re-allocation of N from seed storage proteins toward the hypocotyl to be later used during early development of secondary vascular system.
Collapse
Affiliation(s)
- Rafael A Cañas
- Departamento Biología Molecular y Bioquímica, Universidad de Málaga, Campus de Teatinos s/n, 29071 Málaga, Spain
| | | | | | | |
Collapse
|
46
|
Wang Y, Guo HC. Crystallographic snapshot of a productive glycosylasparaginase-substrate complex. J Mol Biol 2006; 366:82-92. [PMID: 17157318 PMCID: PMC1865511 DOI: 10.1016/j.jmb.2006.09.051] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2006] [Revised: 09/02/2006] [Accepted: 09/20/2006] [Indexed: 11/18/2022]
Abstract
Glycosylasparaginase (GA) plays an important role in asparagine-linked glycoprotein degradation. A deficiency in the activity of human GA leads to a lysosomal storage disease named aspartylglycosaminuria. GA belongs to a superfamily of N-terminal nucleophile hydrolases that autoproteolytically generate their mature enzymes from inactive single chain protein precursors. The side-chain of the newly exposed N-terminal residue then acts as a nucleophile during substrate hydrolysis. By taking advantage of mutant enzyme of Flavobacterium meningosepticum GA with reduced enzymatic activity, we have obtained a crystallographic snapshot of a productive complex with its substrate (NAcGlc-Asn), at 2.0 A resolution. This complex structure provided us an excellent model for the Michaelis complex to examine the specific contacts critical for substrate binding and catalysis. Substrate binding induces a conformational change near the active site of GA. To initiate catalysis, the side-chain of the N-terminal Thr152 is polarized by the free alpha-amino group on the same residue, mediated by the side-chain hydroxyl group of Thr170. Cleavage of the amide bond is then accomplished by a nucleophilic attack at the carbonyl carbon of the amide linkage in the substrate, leading to the formation of an acyl-enzyme intermediate through a negatively charged tetrahedral transition state.
Collapse
Affiliation(s)
| | - Hwai-Chen Guo
- *Corresponding author: Hwai-Chen Guo, Department of Physiology and Biophysics, Boston University School of Medicine, 715 Albany Street, Boston, MA 02118-2526, telephone: 617-638-4023, fax: 617-638-4041, E-mail:
| |
Collapse
|