1
|
Giri A, Hong IS, Kwon TK, Kang JS, Jeong JH, Kweon S, Yook S. Exploring therapeutic and diagnostic potential of cysteine cathepsin as targets for cancer therapy with nanomedicine. Int J Biol Macromol 2025; 315:144324. [PMID: 40398760 DOI: 10.1016/j.ijbiomac.2025.144324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2025] [Revised: 05/05/2025] [Accepted: 05/15/2025] [Indexed: 05/23/2025]
Abstract
Cysteine cathepsins have been discovered to be substantially expressed in multiple types of cancer. They play a key role in the progression and growth of these cancers, rendering them appealing targets for nanoscale delivery and noninvasive diagnostic imaging. This review explores cathepsins from the papain-like enzyme family (C1) within the cysteine peptidase clan (CA), emphasizing the role of cathepsin-responsive nanoparticles in tumor growth. Furthermore, it also explores how nanotechnology can harness cathepsin activity to enable targeted drug delivery, improve tumor imaging, and reduce systemic toxicity. By examining the molecular mechanisms governing cathepsin function and evaluating different nanocarrier systems, this work aims to enhance our understanding of targeted cancer treatment. Despite significant advances, challenges remain in translating these nanomedicine platforms into clinical use, including improving delivery efficiency, biocompatibility, long-term safety, and addressing issues such as interspecies protease variability and scalable nanomanufacturing. Future advancement, integrating advanced biomaterials, patient-derived organoid models, bispecific immune-protease targeting, CRISPR-based cathepsin editing, and artificial intelligence-driven pharmacokinetic modeling and analysis will be critical to fully realizing the clinical potential of cathepsin targeted nanomedicines. These innovations hold promises for advancing precision oncology by overcoming current limitations and improving patient outcomes.
Collapse
Affiliation(s)
- Anil Giri
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - In-Sun Hong
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 406-840, Republic of Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu 42601, Republic of Korea; Center for Forensic Pharmaceutical Science, Keimyung University, Daegu, 42601, Republic of Korea
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jee-Heon Jeong
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon, Gyeonggi 16419, Republic of Korea
| | - Seho Kweon
- College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Simmyung Yook
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea; School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
2
|
Bakkar M, Khalil S, Bhayekar K, Kushwaha ND, Samarbakhsh A, Dorandish S, Edwards H, Dou QP, Ge Y, Gavande NS. Ubiquitin-Specific Protease Inhibitors for Cancer Therapy: Recent Advances and Future Prospects. Biomolecules 2025; 15:240. [PMID: 40001543 PMCID: PMC11853158 DOI: 10.3390/biom15020240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Cancer management has traditionally depended on chemotherapy as the mainstay of treatment; however, recent advancements in targeted therapies and immunotherapies have offered new options. Ubiquitin-specific proteases (USPs) have emerged as promising therapeutic targets in cancer treatment due to their crucial roles in regulating protein homeostasis and various essential cellular processes. This review covers the following: (1) the structural and functional characteristics of USPs, highlighting their involvement in key cancer-related pathways, and (2) the discovery, chemical structures, mechanisms of action, and potential clinical implications of USP inhibitors in cancer therapy. Particular attention is given to the role of USP inhibitors in enhancing cancer immunotherapy, e.g., modulation of the tumor microenvironment, effect on regulatory T cell function, and influence on immune checkpoint pathways. Furthermore, this review summarizes the current progress and challenges of clinical trials involving USP inhibitors as cancer therapy. We also discuss the complexities of achieving target selectivity, the ongoing efforts to develop more specific and potent USP inhibitors, and the potential of USP inhibitors to overcome drug resistance and synergize with existing cancer treatments. We finally provide a perspective on future directions in targeting USPs, including the potential for personalized medicine based on specific gene mutations, underscoring their significant potential for enhancing cancer treatment. By elucidating their mechanisms of action, clinical progress, and potential future applications, we hope that this review could serve as a useful resource for both basic scientists and clinicians in the field of cancer therapeutics.
Collapse
Affiliation(s)
- Mohamad Bakkar
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (EACPHS), Wayne State University, Detroit, MI 48201, USA; (M.B.); (K.B.); (N.D.K.); (A.S.); (S.D.)
- Division of Pediatric Hematology and Oncology, Children’s Hospital of Michigan, Detroit, MI 48201, USA
| | - Sara Khalil
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.K.); (Q.P.D.)
| | - Komal Bhayekar
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (EACPHS), Wayne State University, Detroit, MI 48201, USA; (M.B.); (K.B.); (N.D.K.); (A.S.); (S.D.)
| | - Narva Deshwar Kushwaha
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (EACPHS), Wayne State University, Detroit, MI 48201, USA; (M.B.); (K.B.); (N.D.K.); (A.S.); (S.D.)
| | - Amirreza Samarbakhsh
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (EACPHS), Wayne State University, Detroit, MI 48201, USA; (M.B.); (K.B.); (N.D.K.); (A.S.); (S.D.)
| | - Sadaf Dorandish
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (EACPHS), Wayne State University, Detroit, MI 48201, USA; (M.B.); (K.B.); (N.D.K.); (A.S.); (S.D.)
| | - Holly Edwards
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA;
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute (KCI), Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Q. Ping Dou
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.K.); (Q.P.D.)
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA;
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute (KCI), Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Yubin Ge
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.K.); (Q.P.D.)
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA;
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute (KCI), Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Navnath S. Gavande
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (EACPHS), Wayne State University, Detroit, MI 48201, USA; (M.B.); (K.B.); (N.D.K.); (A.S.); (S.D.)
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute (KCI), Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
3
|
Clark ND, Li C, Malkowski MG. Structural insights into the role of the prosegment binding loop in a papain-superfamily cysteine protease from Treponema denticola. Acta Crystallogr F Struct Biol Commun 2025; 81:53-61. [PMID: 39846228 PMCID: PMC11783177 DOI: 10.1107/s2053230x25000378] [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: 01/15/2025] [Indexed: 01/24/2025] Open
Abstract
Periodontal diseases afflict 20-50% of the global population and carry serious health and economic burdens. Chronic periodontitis is characterized by inflammation of the periodontal pocket caused by dysbiosis. This dysbiosis is coupled with an increase in the population of Treponema denticola, a spirochete bacterium with high mobility and invasivity mediated by a number of virulence factors. One such virulence factor is TDE0362, a multidomain protein with a carboxy-terminal papain-superfamily cysteine protease (C0362). Most papain-superfamily cysteine proteases are produced as proenzymes with a prodomain that interacts with the prosegment binding loop (PBL), requiring proteolytic processing for full activation. Previous studies have indicated that C0362 is not produced as a proenzyme, suggesting an alternative regulatory mechanism. We previously determined the crystal structure of C0362 captured in an inactive conformation with an oxidized catalytic cysteine and a disordered PBL. In this follow-up study, we evaluated the active-site architecture and the PBL in two mutant (Y559A and C412S) structures and an inhibitor-bound (E64) structure to provide insight into the role that the PBL plays in the generation of active enzyme. Our results implicate Tyr559 as playing a critical role in the transition of the enzyme to an active state. We subsequently utilized the structural information to generate models of C0362 bound to human complement factors C3 and C4. Collectively, our results provide insight into the regulatory mechanism and putative substrate-binding interfaces of C0362, highlighting avenues of further research towards inhibition of this essential virulence factor.
Collapse
Affiliation(s)
- N. D. Clark
- Department of Structural Biology, Jacobs School of Medicine and Biomedical SciencesUniversity at Buffalo, State University of New YorkBuffaloNew YorkUSA
| | - C. Li
- Philips Institute for Oral Health Research, School of DentistryVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - M. G. Malkowski
- Department of Structural Biology, Jacobs School of Medicine and Biomedical SciencesUniversity at Buffalo, State University of New YorkBuffaloNew YorkUSA
| |
Collapse
|
4
|
Burroughs AM, Nicastro GG, Aravind L. The Lipocone Superfamily: A Unifying Theme In Metabolism Of Lipids, Peptidoglycan And Exopolysaccharides, Inter-Organismal Conflicts And Immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.14.632903. [PMID: 40236132 PMCID: PMC11996534 DOI: 10.1101/2025.01.14.632903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Wnt proteins are critical signaling molecules in developmental processes across animals. Despite intense study, their evolutionary roots have remained enigmatic. Using sensitive sequence analysis and structure modeling, we establish that the Wnts are part of a vast assemblage of domains, the Lipocone superfamily, defined here for the first time. It includes previously studied enzymatic domains like the phosphatidylserine synthases (PTDSS1/2) and the TelC toxin domain from Streptococcus intermedius , the enigmatic VanZ proteins, the animal Serum Amyloid A (SAA) and a further host of uncharacterized proteins in a total of 30 families. Though the metazoan Wnts are catalytically inactive, we present evidence for a conserved active site across this superfamily, versions of which are consistently predicted to operate on head groups of either phospholipids or polyisoprenoid lipids, catalyzing transesterification and phosphate-containing head group severance reactions. We argue that this superfamily originated as membrane proteins, with one branch (including Wnt and SAA) evolving into soluble versions. By comprehensively analyzing contextual information networks derived from comparative genomics, we establish that they act in varied functional contexts, including regulation of membrane lipid composition, extracellular polysaccharide biosynthesis, and biogenesis of bacterial outer-membrane components, like lipopolysaccharides. On multiple occasions, members of this superfamily, including the bacterial progenitors of Wnt and SAA, have been recruited as effectors in biological conflicts spanning inter-organismal interactions and anti-viral immunity in both prokaryotes and eukaryotes. These findings establish a unifying theme in lipid biochemistry, explain the origins of Wnt signaling and provide new leads regarding immunity across the tree of life.
Collapse
|
5
|
Kusumasari C, Meidyawati R, Megantoro A, Tiara R, Meiskya A, Darwish KM, Abdou A. Development of a novel papain gel formulation: Exploring different concentrations for smear-layer deproteinization and enhanced dentin bonding. Heliyon 2024; 10:e39035. [PMID: 39435069 PMCID: PMC11492583 DOI: 10.1016/j.heliyon.2024.e39035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 10/23/2024] Open
Abstract
Background The self-etch adhesive system modifies but does not completely remove the smear layer, leading to the weakening of the bond strength due to the formation of a hybridized layer. Smear-layer deproteinization with papain enzyme partially removes the smear layer, and increases the bond strength with self-etch adhesive. The aim was to develop a deproteinizing agent with a high papain enzyme concentration to enhance dentin bonding with self-etch adhesives. Methods Papain enzyme gel formulations (15 and 30 IU/g) were prepared and tested for physical stability, viscosity, pH, homogeneity, and organoleptic properties. Moreover, 64 teeth were used to test the deproteinization efficiency of the formed gel. Fourier transform infrared was used to calculate the ratio of organic to inorganic components of smear-layer after deproteinization with 15 and 30 IU/g papain gel and a 6 IU/g commercial papain gel. Moreover, tensile bond strength was measured after deproteinization and dentin bonding with self-etching adhesive for the same groups. A molecular modeling simulation was also performed to evaluate the protein-protein binding interaction, predict the conformational/orientation patterns, and estimate the binding energies of papain with collagen target protein. Results Both 15 and 30 IU/g gels exhibited similar viscosity, pH, homogeneity, and organoleptic properties. However, after 60 s, the 15 IU/g gel was solid, while the 30 IU/g gel was half-solid. All tested groups decreased the amide:phosphate ratio and increased tensile bond strength. Binding complexes between papain and three deposited collagen-1 structures formed strong binding energies with high negative values and residue-wise binding patterns. Conclusions The production of the papain enzyme gel with a concentration of 15 IU/g was successful. In addition, it demonstrated promising results when used as a smear-layer deproteinization agent. Clinical significance Enzymatic smear-layer deproteinization may improve dentin adhesion, and high concertation papain enzyme gels may improve dentin adhesion with the use of self-etch adhesive.
Collapse
Affiliation(s)
- Citra Kusumasari
- Department of Conservative Dentistry, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | - Ratna Meidyawati
- Department of Conservative Dentistry, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | - Aryo Megantoro
- Department of Conservative Dentistry, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | - Rachendra Tiara
- Department of Conservative Dentistry, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | - Agita Meiskya
- Department of Conservative Dentistry, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | - Khaled M. Darwish
- Department of Medicinal Chemistry, Faculty of Pharmacy, Galala University, New Galala, 43713, Egypt
- Department of Medicinal Chemistry, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
| | - Ahmed Abdou
- Department of Restorative Dentistry, Faculty of Dentistry, Universiti Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
6
|
Petruzzella A, Bruand M, Santamaria-Martínez A, Katanayeva N, Reymond L, Wehrle S, Georgeon S, Inel D, van Dalen FJ, Viertl D, Lau K, Pojer F, Schottelius M, Zoete V, Verdoes M, Arber C, Correia BE, Oricchio E. Antibody-peptide conjugates deliver covalent inhibitors blocking oncogenic cathepsins. Nat Chem Biol 2024; 20:1188-1198. [PMID: 38811854 DOI: 10.1038/s41589-024-01627-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 04/18/2024] [Indexed: 05/31/2024]
Abstract
Cysteine cathepsins are a family of proteases that are relevant therapeutic targets for the treatment of different cancers and other diseases. However, no clinically approved drugs for these proteins exist, as their systemic inhibition can induce deleterious side effects. To address this problem, we developed a modular antibody-based platform for targeted drug delivery by conjugating non-natural peptide inhibitors (NNPIs) to antibodies. NNPIs were functionalized with reactive warheads for covalent inhibition, optimized with deep saturation mutagenesis and conjugated to antibodies to enable cell-type-specific delivery. Our antibody-peptide inhibitor conjugates specifically blocked the activity of cathepsins in different cancer cells, as well as osteoclasts, and showed therapeutic efficacy in vitro and in vivo. Overall, our approach allows for the rapid design of selective cathepsin inhibitors and can be generalized to inhibit a broad class of proteases in cancer and other diseases.
Collapse
Affiliation(s)
- Aaron Petruzzella
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
| | - Marine Bruand
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
| | - Albert Santamaria-Martínez
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
| | - Natalya Katanayeva
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
| | - Luc Reymond
- Institute of Chemical Sciences and Engineering (ISIC), Institute of Bioengineering, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Sarah Wehrle
- Laboratory of Protein Design and Immunoengineering, School of Engineering, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Sandrine Georgeon
- Laboratory of Protein Design and Immunoengineering, School of Engineering, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Damla Inel
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Floris J van Dalen
- Department of Medical Biosciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Institute for Chemical Immunology, Nijmegen, The Netherlands
| | - David Viertl
- Translational Radiopharmaceutical Sciences, Departments of Nuclear Medicine and Molecular Imaging and of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
- In Vivo Imaging Facility, Department of Research and Training, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Kelvin Lau
- Protein Production and Structure Core Facility, School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Florence Pojer
- Protein Production and Structure Core Facility, School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Margret Schottelius
- Translational Radiopharmaceutical Sciences, Departments of Nuclear Medicine and Molecular Imaging and of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
- AGORA Pôle de Recherche sur le Cancer, Lausanne, Switzerland
| | - Vincent Zoete
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Martijn Verdoes
- Department of Medical Biosciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Institute for Chemical Immunology, Nijmegen, The Netherlands
| | - Caroline Arber
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Bruno E Correia
- Laboratory of Protein Design and Immunoengineering, School of Engineering, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland.
| | - Elisa Oricchio
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland.
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland.
| |
Collapse
|
7
|
González R, Carvalho ATP. Papain-Mediated Conjugation of Peptide Nucleic Acids to Delivery Peptides: A Density Functional Theory/Molecular Mechanics Metadynamics Study in Aqueous and Organic Solvent. J Phys Chem B 2024; 128:7500-7512. [PMID: 39052428 PMCID: PMC11317979 DOI: 10.1021/acs.jpcb.4c02294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/17/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
Enzymatic peptide synthesis is a powerful alternative to solid-phase methods, as enzymes can have high regio- and stereoselectivity and high yield and require mild reaction conditions. This is beneficial in formulation research due to the rise of nucleic acid therapies. Peptide nucleic acids (PNAs) have a high affinity toward DNA and RNA, and their solubility and cellular delivery can be improved via conjugation to peptides. Here, we designed and assessed the viability of the papain enzyme to conjugate four PNA-peptide models in water and an organic solvent using QM/MM metadynamics. We found that the reactions in water yield better results, where three conjugates could potentially be synthesized by the enzyme, with the first transition state as the rate-limiting step, with an associated energy of 14.53 kcal mol-1, although with a slight endergonic profile. The results highlight the importance of considering the enzyme pockets and different substrate acceptivities and contribute to developing greener, direct, and precise synthetic routes for nucleic acid-based therapies. By exploring the enzyme's potential in conjunction with chemical synthesis, current protocols can be simplified for the synthesis of longer nucleic acids and peptide sequences (and, by extension, proteins) from smaller oligo or peptide blocks.
Collapse
Affiliation(s)
- Ricardo
D. González
- CNC-UC
− Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal
- CIBB
− Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra 3004-504, Portugal
- Institute
for Interdisciplinary Research, Doctoral Programme in Experimental
Biology and Biomedicine (PDBEB), University
of Coimbra, Coimbra 3030-789, Portugal
| | - Alexandra T. P. Carvalho
- CNC-UC
− Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal
- CIBB
− Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra 3004-504, Portugal
- Almac
Sciences, Department of Biocatalysis and Isotope Chemistry, Almac House, 20 Seagoe Industrial Estate ,Craigavon, Northern Ireland BT63 5QD, United Kingdom
| |
Collapse
|
8
|
Soleimani Asl S, Roozbahani MH. A novel robust inhibitor of papain-like protease (PLpro) as a COVID-19 drug. J Biomol Struct Dyn 2024; 42:6863-6870. [PMID: 37578047 DOI: 10.1080/07391102.2023.2245474] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 07/08/2023] [Indexed: 08/15/2023]
Abstract
Regarding the significance of SARS-CoV-2, scientists have shown considerable interest in developing effective drugs. Inhibitors for PLpro are the primary strategies for locating suitable COVID-19 drugs. Natural compounds comprise the majority of COVID-19 drugs. Due to limitations on the safety of clinical trials in cases of COVID, computational methods are typically utilized for inhibition studies. Whereas papain is highly similar to PLpro and is entirely safe, the current study aimed to examine several plant secondary metabolites to identify the most effective papain inhibitor and validate the results using molecular dynamics and docking. This simulation was conducted identically for PLpro and the optimal inhibitor. The results indicated that the experimental results are comparable to those obtained In-Silico, and the inhibition effects of Chlorogenic acid (CGA) on papain attained in the experiment were validated (IC50=0.54 mM). CGA as an inhibitor was located in the active site of PLpro and papain (total energy -2009410 and -456069 kJ/mol, respectively) at the desired location and distance. The study revealed that CGA and its derivatives are effective PLpro inhibitors against SARS-CoV-2.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Saeed Soleimani Asl
- Iran Digital Twin Laboratory (IDT-Lab)- Incubator Center, Iran University of Science and Technology, Tehran, Iran
| | | |
Collapse
|
9
|
Koroleva V, Lavlinskaya M, Holyavka M, Penkov N, Zuev Y, Artyukhov V. Thermal Inactivation, Denaturation and Aggregation Processes of Papain-Like Proteases. Chem Biodivers 2024; 21:e202401038. [PMID: 38849308 DOI: 10.1002/cbdv.202401038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 06/09/2024]
Abstract
The investigation into the behavior of ficin, bromelain, papain under thermal conditions holds both theoretical and practical significance. The production processes of medicines and cosmetics often involve exposure to high temperatures, particularly during the final product sterilization phase. Hence, it's crucial to identify the "critical" temperatures for each component within the mixture for effective technological regulation. In light of this, the objective of this study was to examine the thermal inactivation, aggregation, and denaturation processes of three papain-like proteases: ficin, bromelain, papain. To achieve this goal, the following experiments were conducted: (1) determination of the quantity of inactivated proteases using enzyme kinetics with BAPNA as a substrate; (2) differential scanning calorimetry (DSC); (3) assessment of protein aggregation using dynamic light scattering (DLS) and spectrophotometric analysis at 280 nm. Our findings suggest that the inactivation of ficin and papain exhibits single decay step which characterized by a rapid decline, then preservation of the same residual activity by enzyme stabilization. Only bromelain shows two steps with different kinetics. The molecular sizes of the active and inactive forms are similar across ficin, bromelain, and papain. Furthermore, the denaturation of these forms occurs at approximately the same rate and is accompanied by protein aggregation.
Collapse
Affiliation(s)
- Victoria Koroleva
- Department of Biophysics and Biotechnology, Voronezh State University, Universitetskaya Sq. 1, Voronezh, Russia
- Department of Biology, Voronezh State Medical University named after N.N. Burdenko, Studencheskaya St. 10, Voronezh, Russia
| | - Maria Lavlinskaya
- Department of Biophysics and Biotechnology, Voronezh State University, Universitetskaya Sq. 1, Voronezh, Russia
| | - Marina Holyavka
- Department of Biophysics and Biotechnology, Voronezh State University, Universitetskaya Sq. 1, Voronezh, Russia
| | - Nikita Penkov
- Laboratories of methods of optical-spectral analysis, Institute of Cell Biophysics of the Russian Academy of Sciences, Institutskaya St. 3, Pushchino, Russia
| | - Yuriy Zuev
- Laboratory of Biophysical Chemistry of Nanosystems, FRC Kazan Scientific Center of Russian Academy of Sciences, Kazan Institute of Biochemistry and Biophysics, Kazan, Russia, Lobachevskogo St. 2/31, Kazan, Russia
| | - Valeriy Artyukhov
- Department of Biophysics and Biotechnology, Voronezh State University, Universitetskaya Sq. 1, Voronezh, Russia
| |
Collapse
|
10
|
Huang G, Cong Z, Liu Z, Chen F, Bravo A, Soberón M, Zheng J, Peng D, Sun M. Silencing Ditylenchus destructor cathepsin L-like cysteine protease has negative pleiotropic effect on nematode ontogenesis. Sci Rep 2024; 14:10030. [PMID: 38693283 PMCID: PMC11063044 DOI: 10.1038/s41598-024-60018-5] [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: 01/20/2024] [Accepted: 04/17/2024] [Indexed: 05/03/2024] Open
Abstract
Ditylenchus destructor is a migratory plant-parasitic nematode that severely harms many agriculturally important crops. The control of this pest is difficult, thus efficient strategies for its management in agricultural production are urgently required. Cathepsin L-like cysteine protease (CPL) is one important protease that has been shown to participate in various physiological and pathological processes. Here we decided to characterize the CPL gene (Dd-cpl-1) from D. destructor. Analysis of Dd-cpl-1 gene showed that Dd-cpl-1 gene contains a signal peptide, an I29 inhibitor domain with ERFNIN and GNFD motifs, and a peptidase C1 domain with four conserved active residues, showing evolutionary conservation with other nematode CPLs. RT-qPCR revealed that Dd-cpl-1 gene displayed high expression in third-stage juveniles (J3s) and female adults. In situ hybridization analysis demonstrated that Dd-cpl-1 was expressed in the digestive system and reproductive organs. Silencing Dd-cpl-1 in 1-cell stage eggs of D. destructor by RNAi resulted in a severely delay in development or even in abortive morphogenesis during embryogenesis. The RNAi-mediated silencing of Dd-cpl-1 in J2s and J3s resulted in a developmental arrest phenotype in J3 stage. In addition, silencing Dd-cpl-1 gene expression in female adults led to a 57.43% decrease in egg production. Finally, Dd-cpl-1 RNAi-treated nematodes showed a significant reduction in host colonization and infection. Overall, our results indicate that Dd-CPL-1 plays multiple roles in D. destructor ontogenesis and could serve as a new potential target for controlling D. destructor.
Collapse
Affiliation(s)
- Guoqiang Huang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Ziwen Cong
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Zhonglin Liu
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Feng Chen
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Alejandra Bravo
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, 62210, Cuernavaca, Morelos, Mexico
| | - Mario Soberón
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, 62210, Cuernavaca, Morelos, Mexico
| | - Jinshui Zheng
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Donghai Peng
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Ming Sun
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China.
- Hubei Hongshan Laboratory, Wuhan, 430070, China.
| |
Collapse
|
11
|
Reim T, Ehrt C, Graef J, Günther S, Meents A, Rarey M. SiteMine: Large-scale binding site similarity searching in protein structure databases. Arch Pharm (Weinheim) 2024; 357:e2300661. [PMID: 38335311 DOI: 10.1002/ardp.202300661] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/10/2024] [Accepted: 01/16/2024] [Indexed: 02/12/2024]
Abstract
Drug discovery and design challenges, such as drug repurposing, analyzing protein-ligand and protein-protein complexes, ligand promiscuity studies, or function prediction, can be addressed by protein binding site similarity analysis. Although numerous tools exist, they all have individual strengths and drawbacks with regard to run time, provision of structure superpositions, and applicability to diverse application domains. Here, we introduce SiteMine, an all-in-one database-driven, alignment-providing binding site similarity search tool to tackle the most pressing challenges of binding site comparison. The performance of SiteMine is evaluated on the ProSPECCTs benchmark, showing a promising performance on most of the data sets. The method performs convincingly regarding all quality criteria for reliable binding site comparison, offering a novel state-of-the-art approach for structure-based molecular design based on binding site comparisons. In a SiteMine showcase, we discuss the high structural similarity between cathepsin L and calpain 1 binding sites and give an outlook on the impact of this finding on structure-based drug design. SiteMine is available at https://uhh.de/naomi.
Collapse
Affiliation(s)
- Thorben Reim
- ZBH - Center for Bioinformatics, Universität Hamburg, Hamburg, Germany
| | - Christiane Ehrt
- ZBH - Center for Bioinformatics, Universität Hamburg, Hamburg, Germany
| | - Joel Graef
- ZBH - Center for Bioinformatics, Universität Hamburg, Hamburg, Germany
| | - Sebastian Günther
- Center for Free-Electron Laser Science CFEL, Deutsches Elektronen-Synchrotron DESY, Hamburg, Germany
| | - Alke Meents
- Center for Free-Electron Laser Science CFEL, Deutsches Elektronen-Synchrotron DESY, Hamburg, Germany
| | - Matthias Rarey
- ZBH - Center for Bioinformatics, Universität Hamburg, Hamburg, Germany
| |
Collapse
|
12
|
Duran Ş, Üstüntanir Dede AF, Dündar Orhan Y, Arslanyolu M. Genome-wide identification and in-silico analysis of papain-family cysteine protease encoding genes in Tetrahymena thermophila. Eur J Protistol 2024; 92:126033. [PMID: 38088016 DOI: 10.1016/j.ejop.2023.126033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/06/2023] [Accepted: 11/02/2023] [Indexed: 02/06/2024]
Abstract
Tetrahymena thermophila is a promising host for recombinant protein production, but its utilization in biotechnology is mostly limited due to the presence of intracellular and extracellular papain-family cysteine proteases (PFCPs). In this study, we employed bioinformatics approaches to investigate the T. thermophila PFCP genes and their encoded proteases (TtPFCPs), the most prominent protease family in the genome. Results from the multiple sequence alignment, protein modeling, and conserved motif analyses revealed that all TtPFCPs showed considerably high homology with mammalian cysteine cathepsins and contained conserved amino acid motifs. The total of 121 TtPFCP-encoding genes, 14 of which were classified as non-peptidase homologs, were found. Remaining 107 true TtPFCPs were divided into four distinct subgroups depending on their homology with mammalian lysosomal cathepsins: cathepsin L-like (TtCATLs), cathepsin B-like (TtCATBs), cathepsin C-like (TtCATCs), and cathepsin X-like (TtCATXs) PFCPs. The majority of true TtPFCPs (96 out of the total) were in TtCATL-like peptidase subgroup. Both phylogenetic and chromosomal localization analyses of TtPFCPs supported the hypothesis that TtPFCPs likely evolved through tandem gene duplication events and predominantly accumulated on micronuclear chromosome 5. Additionally, more than half of the identified TtPFCP genes are expressed in considerably low quantities compared to the rest of the TtPFCP genes, which are expressed at a higher level. However, their expression patterns fluctuate based on the stage of the life cycle. In conclusion, this study provides the first comprehensive in-silico analysis of TtPFCP genes and encoded proteases. The results would help designing an effective strategy for protease knockout mutant cell lines to discover biological function and to improve the recombinant protein production in T. thermophila.
Collapse
Affiliation(s)
- Şeyma Duran
- Department of Molecular Biology, Graduate School of Sciences, Eskisehir Technical University, Yunus Emre Campus, Eskişehir 26470, Türkiye.
| | - Ayça Fulya Üstüntanir Dede
- Department of Molecular Biology, Graduate School of Sciences, Eskisehir Technical University, Yunus Emre Campus, Eskişehir 26470, Türkiye.
| | - Yeliz Dündar Orhan
- Department of Advanced Technologies, Graduate School of Sciences, Eskisehir Technical University, Yunus Emre Campus, Eskişehir 26470, Türkiye.
| | - Muhittin Arslanyolu
- Department of Biology, Faculty of Sciences, Eskisehir Technical University, Yunusemre Campus, Eskişehir 26470, Türkiye.
| |
Collapse
|
13
|
Geri A, Massai L, Novinec M, Turel I, Messori L. Reactions of Medicinal Gold Compounds with Cathepsin B Explored through Electrospray Mass Spectrometry Measurements. Chempluschem 2024; 89:e202300321. [PMID: 37930642 DOI: 10.1002/cplu.202300321] [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: 07/03/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/07/2023]
Abstract
Medicinal gold compounds, a novel class of potential anticancer drugs, are believed to produce their pharmacological effects mainly through direct gold binding to protein targets at the level of solvent exposed cysteine (or selenocysteine) residues. We have explored therein the reactions of a panel of seven representative gold compounds with the cysteine protease cathepsin B according to an established ESI MS approach. Detailed information on the mode of protein binding of these gold compounds is gained; notably, quite distinct patterns of cathepsin B metalation have emerged from these studies. It is shown that panel gold compounds interact preferentially, often exclusively, with the free cysteine located in the active site of the enzyme.
Collapse
Affiliation(s)
- Andrea Geri
- Department of Chemistry "Ugo Schiff", Università di Firenze, Via della Lastruccia 3, 50019, Sesto Fiorentino, Italy
| | - Lara Massai
- Department of Chemistry "Ugo Schiff", Università di Firenze, Via della Lastruccia 3, 50019, Sesto Fiorentino, Italy
| | - Marko Novinec
- Faculty of Chemistry and Chemical Technology, Department of Chemistry and Biochemistry, University of Ljubljana, Večna pot 113, 1000, Ljubljana, Slovenia
| | - Iztok Turel
- Faculty of Chemistry and Chemical Technology, Department of Chemistry and Biochemistry, University of Ljubljana, Večna pot 113, 1000, Ljubljana, Slovenia
| | - Luigi Messori
- Department of Chemistry "Ugo Schiff", Università di Firenze, Via della Lastruccia 3, 50019, Sesto Fiorentino, Italy
| |
Collapse
|
14
|
Yan SW, Cheng YK, Lu QQ, Zhang R, Dan Liu R, Long SR, Wang ZQ, Cui J. Characterization of a novel dipeptidyl peptidase 1 of Trichinella spiralis and its participation in larval invasion. Acta Trop 2024; 249:107076. [PMID: 37977254 DOI: 10.1016/j.actatropica.2023.107076] [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: 07/03/2023] [Revised: 10/19/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
The research aimed to describe a new Trichinella spiralis dipeptidyl peptidase 1 (TsDPP1) and investigate its functions in the larval invasion of intestinal epithelial cells (IECs). The gene TsDPP1 was successfully replicated and produced in Escherichia coli BL21 (DE3), showing a strong immune response. TsDPP1 was detected in diverse stages of T. spiralis and showed significant expression in the intestine infective larvae (IIL) and adult worms at 6 days post infection, as confirmed by qPCR and Western blot analysis. The primary localization of TsDPP1 in this parasite was observed in cuticles, stichosomes, and embryos by using the indirect immunofluorescence assay (IIFA). rTsDPP1 exhibited the enzymatic function of natural dipeptidyl peptidase and showed specific binding to IECs, and the binding site was found to be localized on cell membrane. Following transfection with dsRNA-TsDPP1, the expression of TsDPP1 mRNA and protein in muscle larvae (ML) were decreased by approximately 63.52 % and 58.68 %, correspondingly. The activity of TsDPP1 in the ML and IIL treated with dsRNA-TsDPP1 was reduced by 42.98 % and 45.07 %, respectively. The acceleration of larval invasion of IECs was observed with rTsDPP1, while the invasion was suppressed by anti-rTsDPP1 serum. The ability of the larvae treated with dsRNA-TsDPP1 to invade IECs was hindered by 31.23 %. In mice infected with dsRNA-treated ML, the intestinal IIL, and adults experienced a significant decrease in worm burdens and a noticeable reduction in adult female length and fecundity compared to the PBS group. These findings indicated that TsDPP1 significantly impedes the invasion, growth, and reproductive capacity of T. spiralis in intestines, suggesting its potential as a target for anti-Trichinella vaccines.
Collapse
Affiliation(s)
- Shu Wei Yan
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052, PR China
| | - Yong Kang Cheng
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052, PR China
| | - Qi Qi Lu
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052, PR China
| | - Ru Zhang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052, PR China
| | - Ruo Dan Liu
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052, PR China
| | - Shao Rong Long
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052, PR China
| | - Zhong Quan Wang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052, PR China.
| | - Jing Cui
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052, PR China.
| |
Collapse
|
15
|
Zhang Z, Zhan F. Type 2 Cystatins and Their Roles in the Regulation of Human Immune Response and Cancer Progression. Cancers (Basel) 2023; 15:5363. [PMID: 38001623 PMCID: PMC10670837 DOI: 10.3390/cancers15225363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/08/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Cystatins are a family of intracellular and extracellular protease inhibitors that inhibit cysteine cathepsins-a group of lysosomal cysteine proteases that participate in multiple biological processes, including protein degradation and post-translational cleavage. Cysteine cathepsins are associated with the development of autoimmune diseases, tumor progression, and metastasis. Cystatins are categorized into three subfamilies: type 1, type 2, and type 3. The type 2 cystatin subfamily is the largest, containing 10 members, and consists entirely of small secreted proteins. Although type 2 cystatins have many shared biological roles, each member differs in structure, post-translational modifications (e.g., glycosylation), and expression in different cell types. These distinctions allow the type 2 cystatins to have unique biological functions and properties. This review provides an overview of type 2 cystatins, including their biological similarities and differences, their regulatory effect on human immune responses, and their roles in tumor progression, immune evasion, and metastasis.
Collapse
Affiliation(s)
| | - Fenghuang Zhan
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| |
Collapse
|
16
|
Mousavi Ghahfarrokhi SS, Mahdigholi FS, Amin M. Collateral beauty in the damages: an overview of cosmetics and therapeutic applications of microbial proteases. Arch Microbiol 2023; 205:375. [PMID: 37935975 DOI: 10.1007/s00203-023-03713-7] [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: 09/12/2023] [Revised: 10/14/2023] [Accepted: 10/16/2023] [Indexed: 11/09/2023]
Abstract
Microbial proteases are enzymes secreted by a variety of microorganisms, including bacteria and fungi, and have attracted significant attention due to their versatile applications in the food and pharmaceutical industries. In addition, certain proteases have been used in the development of skin health products and cosmetics. This article provides a review of microbial proteases in terms of their classification, sources, properties, and applications. Moreover, different pharmacological and molecular investigations have been reviewed. Various biological activities of microbial proteases, such as Arazyme, collagenase, elastin, and Nattokinase, which are involved in the digestion of dietary proteins, as well as their potential anti-inflammatory, anti-cancer, antithrombotic, and immunomodulatory effects have been included. Furthermore, their ability to control infections and treat various disorders has been discussed. Finally, this review highlights the potential applications and future perspectives of microbial proteases in biotechnology and biomedicine, and proposes further studies to develop new perspectives for disease control and health-promoting strategies using microbial resources.
Collapse
Affiliation(s)
- Seyed Sadeq Mousavi Ghahfarrokhi
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Pharmaceutical Microbiology Group, Pharmaceutical Quality Assurance Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Fateme Sadat Mahdigholi
- Department of Biomaterials, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Amin
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
- Pharmaceutical Microbiology Group, Pharmaceutical Quality Assurance Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.
- Room No. 1-221, Faculty of Pharmacy, 16th Azar Street, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
17
|
Iguchi R, Nakayama S, Sasakura Y, Sekiguchi T, Ogasawara M. Repetitive and zonal expression profiles of absorption-related genes in the gastrointestinal tract of ascidian Ciona intestinalis type A. Cell Tissue Res 2023; 394:343-360. [PMID: 37670165 DOI: 10.1007/s00441-023-03828-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 08/15/2023] [Indexed: 09/07/2023]
Abstract
Intestinal absorption is essential for heterotrophic bilaterians with a tubular gut. Although the fundamental features of the digestive system were shared among chordates with evolution, the gut morphologies of vertebrates diverged and adapted to different food habitats. The ascidian Ciona intestinalis type A, a genome-wide research model of basal chordates, is used to examine the functional morphology of the intestines because of its transparent juvenile body. In the present study, the characteristic gene expression patterns (GEP) of Ciona absorptive proteins, e.g., brush border membrane enzymes for terminal digestion (lactase, maltase, APA, and APN) and transporters (SGLT1, GLUT5, PEPT1, and B0AT1), were investigated in juveniles and young adults, with a special reference to the absorption of other nutrients by pinocytosis- and phagocytosis-related proteins (megalin, cubilin, amnionless, Dab2, Rab7, LAMP, cathepsins, and MRC1). Whole-mount in situ hybridization revealed that these GEP showed multi-regional and repetitive features along the Ciona gastrointestinal tract, mainly in the stomach and several regions of the intestines. In young adults, many absorption-related genes, including pinocytosis-/phagocytosis-related genes, were also expressed between the stomach and mid-intestine. In the gastrointestinal epithelium, absorption-related genes showed zonal GEP along the epithelial structure. Comparisons of GEP, including other intestinal functions, such as nutrient digestion and intestinal protection, indicated the repetitive assignment of a well-coordinated set of intestinal GEP in the Ciona gastrointestinal tract.
Collapse
Affiliation(s)
- Rin Iguchi
- Department of Biology, Graduate School of Science, Chiba University, 1-33 Yayoi-Cho, Inage-Ku, Chiba, 263-8522, Japan
| | - Satoshi Nakayama
- Department of Biology, Graduate School of Science, Chiba University, 1-33 Yayoi-Cho, Inage-Ku, Chiba, 263-8522, Japan
| | - Yasunori Sasakura
- Shimoda Marine Research Center, University of Tsukuba, 5-10-1 Shimoda, Shizuoka, 415-0025, Japan
| | - Toshio Sekiguchi
- The Noto Marine Laboratory, Division of Marine Environmental Studies, Institute of Nature and Environmental Technology, Kanazawa University, Housu-Gun, Ishikawa, 927-0553, Japan
| | - Michio Ogasawara
- Department of Biology, Graduate School of Science, Chiba University, 1-33 Yayoi-Cho, Inage-Ku, Chiba, 263-8522, Japan.
| |
Collapse
|
18
|
Gul I, Abbas MN, Kausar S, Luo J, Gao X, Mu Y, Fan W, Cui H. Insight into crustacean cathepsins: Structure-evolutionary relationships and functional roles in physiological processes. FISH & SHELLFISH IMMUNOLOGY 2023:108852. [PMID: 37295735 DOI: 10.1016/j.fsi.2023.108852] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/21/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023]
Abstract
Cathepsins belong to a group of proteins that are present in both prokaryotic and eukaryotic organisms and have an extremely high degree of evolutionary conservation. These proteins are functionally active in extracellular environments as soluble enzymatic proteins or attached to plasma membrane receptors. In addition, they occur in cellular secretory vesicles, mitochondria, the cytosol, and within the nuclei of eukaryotic cells. Cathepsins are classified into various groups based on their sequence variations, leading to their structural and functional diversification. The molecular understanding of the physiology of crustaceans has shown that proteases, including cathepsins, are expressed ubiquitously. They also contain one of the central regulatory systems for crustacean reproduction, growth, and immune responses. This review focuses on various aspects of the crustaceans cathepsins and emphasizes their biological roles in different physiological processes such as reproduction, growth, development, and immune responses. We also describe the bioactivity of crustaceans cathepsins. Because of the vital biological roles that cathepsins play as cellular proteases in physiological processes, they have been proposed as potential novel targets for the development of management strategies for the aquaculture industries.
Collapse
Affiliation(s)
- Isma Gul
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, 400716, China; State Key Laboratory of Resource Insects, Southwest University, Chongqing, 400716, China
| | - Muhammad Nadeem Abbas
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, 400716, China; State Key Laboratory of Resource Insects, Southwest University, Chongqing, 400716, China
| | - Saima Kausar
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, 400716, China; State Key Laboratory of Resource Insects, Southwest University, Chongqing, 400716, China
| | - Jili Luo
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, 400716, China; State Key Laboratory of Resource Insects, Southwest University, Chongqing, 400716, China
| | - Xinyue Gao
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, 400716, China; State Key Laboratory of Resource Insects, Southwest University, Chongqing, 400716, China
| | - Yuhang Mu
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, 400716, China; State Key Laboratory of Resource Insects, Southwest University, Chongqing, 400716, China
| | - Wenhui Fan
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, 400716, China; Department of Neurology, Chongqing Ninth People's Hospital, Chongqing, 400700, China.
| | - Honghuan Cui
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, 400716, China; State Key Laboratory of Resource Insects, Southwest University, Chongqing, 400716, China.
| |
Collapse
|
19
|
Magoch M, McEwen AG, Napolitano V, Władyka B, Dubin G. Crystal Structure of Staphopain C from Staphylococcus aureus. Molecules 2023; 28:molecules28114407. [PMID: 37298883 DOI: 10.3390/molecules28114407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Staphylococcus aureus is a common opportunistic pathogen of humans and livestock that causes a wide variety of infections. The success of S. aureus as a pathogen depends on the production of an array of virulence factors including cysteine proteases (staphopains)-major secreted proteases of certain strains of the bacterium. Here, we report the three-dimensional structure of staphopain C (ScpA2) of S. aureus, which shows the typical papain-like fold and uncovers a detailed molecular description of the active site. Because the protein is involved in the pathogenesis of a chicken disease, our work provides the foundation for inhibitor design and potential antimicrobial strategies against this pathogen.
Collapse
Affiliation(s)
- Malgorzata Magoch
- Malopolska Centre of Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
| | - Alastair G McEwen
- CNRS, INSERM, Université de Strasbourg, IGBMC UMR 7104-UMR-S 1258, F-67400 Illkirch, France
| | - Valeria Napolitano
- Malopolska Centre of Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
| | - Benedykt Władyka
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
| | - Grzegorz Dubin
- Malopolska Centre of Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
| |
Collapse
|
20
|
Jeong HW, Chang DS, Kim JS, Hwang YS. Role of cathepsin D induced by Porphyromonas gingivalis lipopolysaccharide in periodontitis. Eur J Oral Sci 2023; 131:e12923. [PMID: 36788303 DOI: 10.1111/eos.12923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 01/26/2023] [Indexed: 02/16/2023]
Abstract
Periodontitis is an inflammatory disease of tooth-supporting tissues caused by oral bacteria. Periodontal ligament loss and alveolar bone destruction occur in progressive periodontitis. Since gingival crevicular fluids (GCF) reflects the inflammatory environment of the periodontal pocket, it is a very important specimen for developing targets for periodontitis diagnosis. An antibody array was performed using GCF collected from healthy participants and patients with periodontitis to identify the proteolytic enzymes involved in periodontitis. Of 21 targets on the antibody array membrane, kallikrein 6 (KLK6), kallikrein 10 (KLK10), cathepsin A (CathA), and cathepsin D (CathD) showed higher levels in periodontitis GCF than in GCF from healthy participants. Lipopolysaccharide stimulation of Porphyromonas gingivalis (PG-LPS) in immortalized gingival fibroblasts only increased CathD protein levels among the four targets. The substrate cleavage activity of CathD was increased in PG-LPS-treated immortalized gingival fibroblast extract. The PG-LPS-induced substrate cleavage effect was abolished by the CathD inhibitor pepstatin A. Osteoclast formation was promoted by treatment with conditioned media from PG-LPS- treated immortalized gingival fibroblasts but inhibited by the CathD inhibitor pepstatin A. These results suggest that PG-LPS affected the osteoclast formation process by increasing CathD expression in cells around the alveolar bone, thereby participating in periodontitis progression.
Collapse
Affiliation(s)
- Hyun Woong Jeong
- Department of Dental Hygiene, College of Health, Science, Eulji University, Seongnam, Korea
| | - Dong Sik Chang
- Department of Otorhinolaryngology, Eulji University Hospital, Eulji University, Daejeon, Korea
| | | | - Young Sun Hwang
- Department of Dental Hygiene, College of Health, Science, Eulji University, Seongnam, Korea
| |
Collapse
|
21
|
Chauke E, Pelle R, Coetzer THT. A single exon-encoded Theileria parva strain Muguga cysteine protease (ThpCP): Molecular modelling and characterisation. Biochimie 2023; 206:24-35. [PMID: 36198333 DOI: 10.1016/j.biochi.2022.09.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/01/2022] [Accepted: 09/26/2022] [Indexed: 11/27/2022]
Abstract
The tick-transmitted apicomplexan Theileria parva causes East Coast fever, a bovine disease of great economic and veterinary importance in Africa. Papain-like cysteine proteases play important roles in protozoan parasite host cell entry and egress, nutrition and host immune evasion. This study reports the identification and characterisation of a T. parva strain Muguga cathepsin L-like (C1A subfamily) cysteine protease (ThpCP). Molecular modelling confirmed the papain-like fold of ThpCP, hydrophobic character of the S2 substrate binding pocket and non-covalent interaction between the pro- and catalytic domains preceding low pH autoactivation. ThpCP was recombinantly expressed in a protease deficient E. coli (Rosetta (DE3)pLysS strain) expression host as a 46 kDa proenzyme. Following Ni-chelate affinity chromatography and acidification, the 27 kDa mature ThpCP was purified by cation-exchange chromatography. Purified ThpCP hydrolysed typical cathepsin L substrates N-α-benzyloxycarbonyl (Z)-Phe-Arg-7-amino-4-methyl-coumarin (AMC) (kcat/Km = 4.49 × 105 s-1M-1) and Z-Leu-Arg-AMC (kcat/Km = 4.20 × 105 s-1M-1), but showed no activity against the cathepsin B-selective substrate Z-Arg-Arg-AMC. Recombinant ThpCP was active over a broad pH range from pH 4.5 to 7.5, thereby showing potential activity in the acidic parasite food vacuole and close to neutral pH of the host lymphocyte cytoplasm. Recombinant ThpCP was inhibited by the cysteine protease inhibitors E64, iodoacetate, leupeptin, chymostatin, Z-Phe-Ala-diazomethylketone (DMK) and Z-Phe-Phe-DMK and hydrolysed bovine proteins: haemoglobin, immunoglobulin G, serum albumin and fibrinogen as well as goat IgG at pH 6 and 7. Functional expression and characterisation of Theileria cysteine proteases should enable high throughput screening of cysteine protease inhibitor libraries against these proteases.
Collapse
Affiliation(s)
- Ephraim Chauke
- Biochemistry, School of Life Sciences, University of KwaZulu-Natal (Pietermaritzburg Campus), Private Bag X01, Scottsville, 3209, South Africa
| | - Roger Pelle
- Biosciences Eastern and Central Africa, International Livestock Research Institute (BecA-ILRI) Hub, P.O. Box, 30709-00100, Nairobi, Kenya
| | - Theresa H T Coetzer
- Biochemistry, School of Life Sciences, University of KwaZulu-Natal (Pietermaritzburg Campus), Private Bag X01, Scottsville, 3209, South Africa.
| |
Collapse
|
22
|
Sorokin AV, Goncharova SS, Lavlinskaya MS, Holyavka MG, Faizullin DA, Kondratyev MS, Kannykin SV, Zuev YF, Artyukhov VG. Carboxymethyl Cellulose-Based Polymers as Promising Matrices for Ficin Immobilization. Polymers (Basel) 2023; 15:polym15030649. [PMID: 36771951 PMCID: PMC9920955 DOI: 10.3390/polym15030649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/20/2023] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
The present work is devoted to research on the interaction between carboxymethyl cellulose sodium salt and its derivatives (graft copolymer of carboxymethyl cellulose sodium salt and N,N-dimethyl aminoethyl methacrylate) with cysteine protease (ficin). The interaction was studied by FTIR and by flexible molecular docking, which have shown the conjugates' formation with both matrices. The proteolytic activity assay performed with azocasein demonstrated that the specific activities of all immobilized ficin samples are higher in comparison with those of the native enzyme. This is due to the modulation of the conformation of ficin globule and of the enzyme active site by weak physical interactions involving catalytically valuable amino acids. The results obtained can extend the practical use of ficin in biomedicine and biotechnology.
Collapse
Affiliation(s)
- Andrey V. Sorokin
- Biophysics and Biotechnology Department, Voronezh State University, 1 Universitetskaya Square, 394018 Voronezh, Russia
- Laboratory of Bioresource Potential of Coastal Area, Institute for Advanced Studies, Sevastopol State University, 33 Studencheskaya Street, 299053 Sevastopol, Russia
- Laboratory of Metagenomics and Food Biotechnologies, Voronezh State University of Engineering Technologies, 19 Revolutsii Avenue, 394036 Voronezh, Russia
| | - Svetlana S. Goncharova
- Biophysics and Biotechnology Department, Voronezh State University, 1 Universitetskaya Square, 394018 Voronezh, Russia
| | - Maria S. Lavlinskaya
- Biophysics and Biotechnology Department, Voronezh State University, 1 Universitetskaya Square, 394018 Voronezh, Russia
- Laboratory of Bioresource Potential of Coastal Area, Institute for Advanced Studies, Sevastopol State University, 33 Studencheskaya Street, 299053 Sevastopol, Russia
- Laboratory of Metagenomics and Food Biotechnologies, Voronezh State University of Engineering Technologies, 19 Revolutsii Avenue, 394036 Voronezh, Russia
| | - Marina G. Holyavka
- Biophysics and Biotechnology Department, Voronezh State University, 1 Universitetskaya Square, 394018 Voronezh, Russia
- Laboratory of Bioresource Potential of Coastal Area, Institute for Advanced Studies, Sevastopol State University, 33 Studencheskaya Street, 299053 Sevastopol, Russia
| | - Dzhigangir A. Faizullin
- Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center of the RAS, 2/31 Lobachevsky Street, 420111 Kazan, Russia
| | - Maxim S. Kondratyev
- Biophysics and Biotechnology Department, Voronezh State University, 1 Universitetskaya Square, 394018 Voronezh, Russia
- Laboratory of Structure and Dynamics of Biomolecular Systems, Institute of Cell Biophysics of the RAS, 3 Institutskaya Street, 142290 Pushchino, Russia
| | - Sergey V. Kannykin
- Material Science and Nanosystem Industry Department, Voronezh State University, 1 Universitetskaya Square, 394018 Voronezh, Russia
| | - Yuriy F. Zuev
- Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center of the RAS, 2/31 Lobachevsky Street, 420111 Kazan, Russia
- Correspondence:
| | - Valeriy G. Artyukhov
- Biophysics and Biotechnology Department, Voronezh State University, 1 Universitetskaya Square, 394018 Voronezh, Russia
| |
Collapse
|
23
|
Puja S, Seth S, Hora R, Kaur S, Mishra PC. Understanding the Molecular Basis for Enhanced Glutenase Activity of Actinidin using Structural Bioinformatics. Protein Pept Lett 2023; 30:777-782. [PMID: 37592795 DOI: 10.2174/0929866530666230817141100] [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: 02/27/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 08/19/2023]
Abstract
BACKGROUND Management of gluten intolerance is currently possible only by consumption of a gluten-free diet (GFD) for a lifetime. The scientific community has been searching for alternatives to GFD, like the inclusion of natural proteases with meals or pre-treatment of gluten-containing foods with glutenases. Actinidin from kiwifruit has shown considerable promise in digesting immunogenic gliadin peptides compared to other plant-derived cysteine proteases. METHODS In this study, we aimed to understand the structural basis for the elevated protease action of actinidin against gliadin peptides by using an in silico approach. RESULTS Docking experiments revealed key differences between the binding of gliadin peptide to actinidin and papain, which may be responsible for their differential digestive action. CONCLUSION Sequence comparison of different plant cysteine proteases highlights amino acid residues surrounding the active site pocket of actinidin that are unique to this molecule and hence likely to contribute to its digestive properties.
Collapse
Affiliation(s)
- Shivangi Puja
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, Punjab-143005, India
| | - Shreya Seth
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, Punjab-143005, India
| | - Rachna Hora
- Department of Molecular Biology and Biochemistry, Guru Nanak Dev University, Amritsar, Punjab-143005, India
| | - Satinder Kaur
- Department of Molecular Biology and Biochemistry, Guru Nanak Dev University, Amritsar, Punjab-143005, India
| | | |
Collapse
|
24
|
Holyavka MG, Goncharova SS, Sorokin AV, Lavlinskaya MS, Redko YA, Faizullin DA, Baidamshina DR, Zuev YF, Kondratyev MS, Kayumov AR, Artyukhov VG. Novel Biocatalysts Based on Bromelain Immobilized on Functionalized Chitosans and Research on Their Structural Features. Polymers (Basel) 2022; 14:5110. [PMID: 36501516 PMCID: PMC9739615 DOI: 10.3390/polym14235110] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022] Open
Abstract
Enzyme immobilization on various carriers represents an effective approach to improve their stability, reusability, and even change their catalytic properties. Here, we show the mechanism of interaction of cysteine protease bromelain with the water-soluble derivatives of chitosan-carboxymethylchitosan, N-(2-hydroxypropyl)-3-trimethylammonium chitosan, chitosan sulfate, and chitosan acetate-during immobilization and characterize the structural features and catalytic properties of obtained complexes. Chitosan sulfate and carboxymethylchitosan form the highest number of hydrogen bonds with bromelain in comparison with chitosan acetate and N-(2-hydroxypropyl)-3-trimethylammonium chitosan, leading to a higher yield of protein immobilization on chitosan sulfate and carboxymethylchitosan (up to 58 and 65%, respectively). In addition, all derivatives of chitosan studied in this work form hydrogen bonds with His158 located in the active site of bromelain (except N-(2-hydroxypropyl)-3-trimethylammonium chitosan), apparently explaining a significant decrease in the activity of biocatalysts. The N-(2-hydroxypropyl)-3-trimethylammonium chitosan displays only physical interactions with His158, thus possibly modulating the structure of the bromelain active site and leading to the hyperactivation of the enzyme, up to 208% of the total activity and 158% of the specific activity. The FTIR analysis revealed that interaction between N-(2-hydroxypropyl)-3-trimethylammonium chitosan and bromelain did not significantly change the enzyme structure. Perhaps this is due to the slowing down of aggregation and the autolysis processes during the complex formation of bromelain with a carrier, with a minimal modification of enzyme structure and its active site orientation.
Collapse
Affiliation(s)
- Marina G. Holyavka
- Biophysics and Biotechnology Department, Voronezh State University, 1 Universitetskaya Square, 394018 Voronezh, Russia
- Laboratory of Bioresource Potential of Coastal Area, Institute for Advanced Studies, Sevastopol State University, 33 Studencheskaya Street, 299053 Sevastopol, Russia
| | - Svetlana S. Goncharova
- Biophysics and Biotechnology Department, Voronezh State University, 1 Universitetskaya Square, 394018 Voronezh, Russia
| | - Andrey V. Sorokin
- Biophysics and Biotechnology Department, Voronezh State University, 1 Universitetskaya Square, 394018 Voronezh, Russia
- Laboratory of Bioresource Potential of Coastal Area, Institute for Advanced Studies, Sevastopol State University, 33 Studencheskaya Street, 299053 Sevastopol, Russia
- Metagenomics and Food Biotechnologies Laboratory, Voronezh State University of Engineering Technologies, 19 Revolutsii Avenue, 394036 Voronezh, Russia
| | - Maria S. Lavlinskaya
- Biophysics and Biotechnology Department, Voronezh State University, 1 Universitetskaya Square, 394018 Voronezh, Russia
- Laboratory of Bioresource Potential of Coastal Area, Institute for Advanced Studies, Sevastopol State University, 33 Studencheskaya Street, 299053 Sevastopol, Russia
- Metagenomics and Food Biotechnologies Laboratory, Voronezh State University of Engineering Technologies, 19 Revolutsii Avenue, 394036 Voronezh, Russia
| | - Yulia A. Redko
- Biophysics and Biotechnology Department, Voronezh State University, 1 Universitetskaya Square, 394018 Voronezh, Russia
| | - Dzhigangir A. Faizullin
- Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center of the RAS, 2/31 Lobachevsky Street, 420111 Kazan, Russia
| | - Diana R. Baidamshina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlevskaya Street, 420008 Kazan, Russia
| | - Yuriy F. Zuev
- Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center of the RAS, 2/31 Lobachevsky Street, 420111 Kazan, Russia
| | - Maxim S. Kondratyev
- Biophysics and Biotechnology Department, Voronezh State University, 1 Universitetskaya Square, 394018 Voronezh, Russia
- Laboratory of Structure and Dynamics of Biomolecular Systems, Institute of Cell Biophysics of the RAS, 3 Institutskaya Street, 142290 Pushchino, Russia
| | - Airat R. Kayumov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlevskaya Street, 420008 Kazan, Russia
| | - Valeriy G. Artyukhov
- Biophysics and Biotechnology Department, Voronezh State University, 1 Universitetskaya Square, 394018 Voronezh, Russia
| |
Collapse
|
25
|
Structure determinants defining the specificity of papain-like cysteine proteases. Comput Struct Biotechnol J 2022; 20:6552-6569. [DOI: 10.1016/j.csbj.2022.11.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/19/2022] [Accepted: 11/20/2022] [Indexed: 11/25/2022] Open
|
26
|
Kinetic Characterization of Cerium and Gallium Ions as Inhibitors of Cysteine Cathepsins L, K, and S. Int J Mol Sci 2022; 23:ijms23168993. [PMID: 36012257 PMCID: PMC9409168 DOI: 10.3390/ijms23168993] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 11/24/2022] Open
Abstract
Heavy metal ions can disrupt biological functions via multiple molecular mechanisms, including inhibition of enzymes. We investigate the interactions of human papain-like cysteine endopeptidases cathepsins L, K, and S with gallium and cerium ions, which are associated with medical applications. We compare these results with zinc and lead, which are known to inhibit thiol enzymes. We show that Ga3+, Ce3+, and Ce4+ ions inhibit all tested peptidases with inhibition constants in the low micromolar range (between 0.5 µM and 10 µM) which is comparable to Zn2+ ions, whereas inhibition constants of Pb2+ ions are one order of magnitude higher (30 µM to 150 µM). All tested ions are linear specific inhibitors of cathepsin L, but cathepsins K and S are inhibited by Ga3+, Ce3+, and Ce4+ ions via hyperbolic inhibition mechanisms. This indicates a mode of interaction different from that of Zn2+ and Pb2+ ions, which act as linear specific inhibitors of all peptidases. All ions also inhibit the degradation of insoluble elastin, which is a common target of these peptidases in various inflammatory diseases. Our results suggest that these ions and their compounds have the potential to be used as cysteine cathepsin inhibitors in vitro and possibly in vivo.
Collapse
|
27
|
Holyavka MG, Pankova SM, Vyshkvorkina YM, Lukin AN, Kondratyev MS, Artyukhov VG. Chitosan as Photo Protector for Papain Immobilized on Its Matrix. Biophysics (Nagoya-shi) 2022. [DOI: 10.1134/s0006350922030095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
28
|
Jung S, Fuchs N, Grathwol C, Hellmich UA, Wagner A, Diehl E, Willmes T, Sotriffer C, Schirmeister T. New peptidomimetic rhodesain inhibitors with improved selectivity towards human cathepsins. Eur J Med Chem 2022; 238:114460. [PMID: 35597010 DOI: 10.1016/j.ejmech.2022.114460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/10/2022] [Accepted: 05/10/2022] [Indexed: 11/04/2022]
Abstract
Parasitic cysteine proteases such as rhodesain (TbCatL) from Trypanosoma brucei rhodesiense are relevant targets for developing new potential drugs against parasitic diseases (e. g. Human African Trypanosomiasis). Designing selective inhibitors for parasitic cathepsins can be challenging as they share high structural similarities with human cathepsins. In this paper, we describe the development of novel peptidomimetic rhodesain inhibitors by applying a structure-based de novo design approach and molecular docking protocols. The inhibitors with a new scaffold in P2 and P3 position display high selectivity towards trypanosomal rhodesain over human cathepsins L and B and high antitrypanosomal activity. Vinylsulfonate 2a has emerged as a potent rhodesain inhibitor (k2nd = 883 • 103 M-1 s-1) with single-digit nanomolar binding affinity (Ki = 9 nM) and more than 150-fold selectivity towards human cathepsins and it thus constitutes an interesting starting compound for the further development of selective drugs against Human African Trypanosomiasis.
Collapse
Affiliation(s)
- Sascha Jung
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudingerweg 5, Mainz, 55128, Germany
| | - Natalie Fuchs
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudingerweg 5, Mainz, 55128, Germany
| | - Christoph Grathwol
- Institute of Organic Chemistry, Karlsruhe Institute of Technology, Fritz-Haber-Weg 6, Karlsruhe, 76131, Germany
| | - Ute A Hellmich
- Department of Chemistry, Biochemistry Section, Johannes Gutenberg University, Hanns-Dieter-Hüsch-Weg 17, Mainz, 55128, Germany; Center for Biomolecular Magnetic Resonance (BMRZ), Goethe University, Max-von-Laue-Str. 9, Frankfurt, 60438, Germany
| | - Annika Wagner
- Department of Chemistry, Biochemistry Section, Johannes Gutenberg University, Hanns-Dieter-Hüsch-Weg 17, Mainz, 55128, Germany
| | - Erika Diehl
- Department of Chemistry, Biochemistry Section, Johannes Gutenberg University, Hanns-Dieter-Hüsch-Weg 17, Mainz, 55128, Germany
| | - Thomas Willmes
- Institute of Pharmacy and Food Chemistry, University Würzburg, Am Hubland, Würzburg, 97074, Germany
| | - Christoph Sotriffer
- Institute of Pharmacy and Food Chemistry, University Würzburg, Am Hubland, Würzburg, 97074, Germany
| | - Tanja Schirmeister
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudingerweg 5, Mainz, 55128, Germany.
| |
Collapse
|
29
|
The Genome of Rhyzopertha dominica (Fab.) (Coleoptera: Bostrichidae): Adaptation for Success. Genes (Basel) 2022; 13:genes13030446. [PMID: 35328000 PMCID: PMC8956072 DOI: 10.3390/genes13030446] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/16/2022] [Accepted: 02/21/2022] [Indexed: 12/15/2022] Open
Abstract
The lesser grain borer, Rhyzopertha dominica (F.) (Coleoptera: Bostrichidae), is a major global pest of cereal grains. Infestations are difficult to control as larvae feed inside grain kernels, and many populations are resistant to both contact insecticides and fumigants. We sequenced the genome of R. dominica to identify genes responsible for important biological functions and develop more targeted and efficacious management strategies. The genome was assembled from long read sequencing and long-range scaffolding technologies. The genome assembly is 479.1 Mb, close to the predicted genome size of 480.4 Mb by flow cytometry. This assembly is among the most contiguous beetle assemblies published to date, with 139 scaffolds, an N50 of 53.6 Mb, and L50 of 4, indicating chromosome-scale scaffolds. Predicted genes from biologically relevant groups were manually annotated using transcriptome data from adults and different larval tissues to guide annotation. The expansion of carbohydrase and serine peptidase genes suggest that they combine to enable efficient digestion of cereal proteins. A reduction in the copy number of several detoxification gene families relative to other coleopterans may reflect the low selective pressure on these genes in an insect that spends most of its life feeding internally. Chemoreceptor genes contain elevated numbers of pseudogenes for odorant receptors that also may be related to the recent ontogenetic shift of R. dominica to a diet consisting primarily of stored grains. Analysis of repetitive sequences will further define the evolution of bostrichid beetles compared to other species. The data overall contribute significantly to coleopteran genetic research.
Collapse
|
30
|
Varda N, Novinec M. Evolutionary Analysis of Dipeptidyl Peptidase I. Int J Mol Sci 2022; 23:ijms23031852. [PMID: 35163774 PMCID: PMC8836896 DOI: 10.3390/ijms23031852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/01/2022] [Accepted: 02/04/2022] [Indexed: 12/10/2022] Open
Abstract
Human dipeptidyl peptidase I (DPPI) belongs to the family of papain-like cysteine peptidases. Its distinctive features are the unique exclusion domain which enables the eponymous activity and homotetramerization of DPPI, and its dependence on chloride ions for enzymatic activity. The oligomeric state of DPPI is unique in this family of predominantly monomeric peptidases. However, a distant DPPI ortholog from Plasmodium falciparum has been shown to be monomeric, indicating that the oligomeric state of DPPI varies between lineages. The aim of this work was to study the evolution of DPPI, with particular attention to the structural features that determine its characteristic enzymatic activity and preferences, and to reconstruct the evolution of its oligomerization. We analyzed fifty-seven selected sequences of DPPI and confirmed its presence in three lineages, namely, Amorphea (including animals and Amoebozoa), Alveolates and the metamonad Giardia. The amino acid residues that bind the chloride ion are highly conserved in all species, indicating that the dependence on chloride ions for activity is an evolutionarily conserved feature of DPPI. The number of N-glycosylation sites is significantly increased in animals, particularly vertebrates. Analysis of homology models and subunit contacts suggests that oligomerization is likely restricted to DPPIs in the Amorphea group.
Collapse
|
31
|
Dvoryakova EA, Vinokurov KS, Tereshchenkova VF, Dunaevsky YE, Belozersky MA, Oppert B, Filippova IY, Elpidina EN. Primary digestive cathepsins L of Tribolium castaneum larvae: Proteomic identification, properties, comparison with human lysosomal cathepsin L. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2022; 140:103679. [PMID: 34763092 DOI: 10.1016/j.ibmb.2021.103679] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 06/13/2023]
Abstract
We previously described the most highly expressed enzymes from the gut of the red flour beetle, Tribolium castaneum, as cathepsins L. In the present study, two C1 family-specific cysteine cathepsin L enzymes from the larval midgut were isolated and identified using MALDI-TOF MS analysis. The isolated T. castaneum cathepsins were characterized according to their specificity against chromogenic and fluorogenic peptide substrates, and the most efficiently hydrolyzed substrate was Z-FR-pNA with Arg in the P1 subsite. The specificity of insect digestive cathepsins was compared with human lysosomal cathepsin L, the well-studied peptidase of the C1 family cathepsins. T. castaneum digestive cathepsins efficiently hydrolyzed substrates with small and uncharged amino acid residues at P1 (Ala, Gln) more than human cathepsin L. In particular, these insect digestive cathepsins cleaved with higher efficiency the analogs of immunogenic peptides of gliadins, which contribute to autoimmune celiac disease in susceptible people, and thus insect enzymes may be useful in enzymatic treatments for this disease. A bioinformatic study supported by the proteomic analysis of the primary structures of the isolated cathepsins was used to compare tertiary models. The phylogenetic analysis of coleopteran and human cathepsins from the L subfamily indicated that insect digestive cathepsins grouped separately from lysosomal cathepsins.
Collapse
Affiliation(s)
- E A Dvoryakova
- A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, 119991, Russia
| | - K S Vinokurov
- Biology Centre of the Czech Academy of Sciences, Institute of Entomology, Czech Republic, Branišovská 1160/31, České Budějovice, 370 05, Czech Republic
| | - V F Tereshchenkova
- Department of Chemistry, Moscow State University, Moscow, 119991, Russia
| | - Y E Dunaevsky
- A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, 119991, Russia
| | - M A Belozersky
- A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, 119991, Russia
| | - B Oppert
- USDA Agricultural Research Service, Center for Grain and Animal Health Research, Manhattan, KS, 66502, USA.
| | - I Y Filippova
- Department of Chemistry, Moscow State University, Moscow, 119991, Russia
| | - E N Elpidina
- A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
32
|
Soares FEDF, Abreu MADF. Plant cysteine proteases: Potential therapeutic agents against SARS-CoV-2. Phytother Res 2021; 36:3-4. [PMID: 34894014 DOI: 10.1002/ptr.7351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 11/09/2022]
|
33
|
Yoo Y, Choi E, Kim Y, Cha Y, Um E, Kim Y, Kim Y, Lee YS. Therapeutic potential of targeting cathepsin S in pulmonary fibrosis. Biomed Pharmacother 2021; 145:112245. [PMID: 34772578 DOI: 10.1016/j.biopha.2021.112245] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/18/2021] [Accepted: 09/22/2021] [Indexed: 11/16/2022] Open
Abstract
Cathepsin S (CTSS), a lysosomal protease, belongs to a family of cysteine cathepsin proteases that promote degradation of damaged proteins in the endolysosomal pathway. Aberrant CTSS expression and regulation are associated with the pathogenesis of several diseases, including lung diseases. CTSS overexpression causes a variety of pathological processes, including pulmonary fibrosis, with increased CTSS secretion and accelerated extracellular matrix remodeling. Compared to many other cysteine cathepsin family members, CTSS has unique features that it presents limited tissue expression and retains its enzymatic activity at a neutral pH, suggesting its decisive involvement in disease microenvironments. In this review, we investigated the role of CTSS in lung disease, exploring recent studies that have indicated that CTSS mediates fibrosis in unique ways, along with its structure, substrates, and distinct regulation. We also outlined examples of CTSS inhibitors in clinical and preclinical development and proposed CTSS as a potential therapeutic target for pulmonary fibrosis.
Collapse
Affiliation(s)
- YoungJo Yoo
- Graduate School of Pharmaceutical Sciences and College of Pharmacy, Ewha Womans University, Seoul 120-720, Republic of Korea
| | - Eun Choi
- Graduate School of Pharmaceutical Sciences and College of Pharmacy, Ewha Womans University, Seoul 120-720, Republic of Korea
| | - Yejin Kim
- Graduate School of Pharmaceutical Sciences and College of Pharmacy, Ewha Womans University, Seoul 120-720, Republic of Korea
| | - Yunyoung Cha
- Graduate School of Pharmaceutical Sciences and College of Pharmacy, Ewha Womans University, Seoul 120-720, Republic of Korea
| | - Eunhye Um
- Graduate School of Pharmaceutical Sciences and College of Pharmacy, Ewha Womans University, Seoul 120-720, Republic of Korea
| | - Younghwa Kim
- Graduate School of Pharmaceutical Sciences and College of Pharmacy, Ewha Womans University, Seoul 120-720, Republic of Korea
| | - Yunji Kim
- Graduate School of Pharmaceutical Sciences and College of Pharmacy, Ewha Womans University, Seoul 120-720, Republic of Korea
| | - Yun-Sil Lee
- Graduate School of Pharmaceutical Sciences and College of Pharmacy, Ewha Womans University, Seoul 120-720, Republic of Korea.
| |
Collapse
|
34
|
Kang J, Gong P, Ge M, Sadeghnezhad E, Liu Z, Zhang M, Shangguan L, Fang J. "The PLCP gene family of grapevine (Vitis vinifera L.): characterization and differential expression in response to Plasmopara Viticola". BMC PLANT BIOLOGY 2021; 21:499. [PMID: 34717550 PMCID: PMC8556938 DOI: 10.1186/s12870-021-03279-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 10/19/2021] [Indexed: 06/01/2023]
Abstract
BACKGROUND Papain-like cysteine proteases (PLCPs), a large group of cysteine proteases, are structurally related to papain. The members belonging to PLCPs family contribute to plant immunity, senescence, and defense responses in plants. The PLCP gene family has been identified in Arabidopsis, rice, soybean, and cotton. However, no systematic analysis of PLCP genes has been undertaken in grapevine. Since Plasmopara viticola as a destructive pathogen could affect immunity of grapes in the field, we considered that the members belonged to PLCPs family could play a crucial role in defensive mechanisms or programmed cell death. We aimed to evaluate the role of PLCPs in 2 different varieties of grapevines and compared the changes of their expressions with the transcriptional data in response to P. viticola. RESULTS In this study, 23 grapevine PLCP (VvPLCP) genes were identified by comprehensive bioinformatics analysis. Subsequently, the chromosomal localizations, gene structure, conserved domains, phylogenetic relationship, gene duplication, and cis-acting elements were analyzed. Numerous cis-acting elements related to plant development, hormone, and stress responses were identified in the promoter of the VvPLCP genes. Phylogenetic analysis grouped the VvPLCP genes into nine subgroups. The transcription of VvPLCP in different inoculation time points and varieties indicated that VvPLCP may have vital functions in grapevine defense against Plasmopara viticola. According to transcriptome data and qPCR analysis, we observed the increasing expression levels of VvRD21-1 at 72 h after inoculation in resistant variety, inferring that it was related to grape downy mildew resistance. Meanwhile, 3 genes including VvXBCP1, VvSAG12-1, and VvALP1 showed higher expression at 24 h after pathogen inoculation in the susceptible variety and might be related to the downy mildew phenotype. We nominated these four genes to function during hypersensitive response (HR) process, inferring that these genes could be associated with downy mildew resistance in grapes. CONCLUSIONS Our results provide the reference for functional studies of PLCP gene family, and highlight its functions in grapevine defense against P. viticola. The results help us to better understand the complexity of the PLCP gene family in plant immunity and provide valuable information for future functional characterization of specific genes in grapevine.
Collapse
Affiliation(s)
- Jun Kang
- Department of Horticulture, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Peijie Gong
- Department of Horticulture, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China.
| | - Mengqing Ge
- Department of Horticulture, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Ehsan Sadeghnezhad
- Department of Horticulture, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Zhongjie Liu
- Department of Horticulture, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Mengwei Zhang
- Department of Horticulture, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Lingfei Shangguan
- Department of Horticulture, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Jinggui Fang
- Department of Horticulture, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China.
| |
Collapse
|
35
|
Goričan T, Ciber L, Petek N, Svete J, Novinec M. Synthesis and kinetic characterization of hyperbolic inhibitors of human cathepsins K and S based on a succinimide scaffold. Bioorg Chem 2021; 115:105213. [PMID: 34364050 DOI: 10.1016/j.bioorg.2021.105213] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/20/2021] [Accepted: 07/24/2021] [Indexed: 02/03/2023]
Abstract
Cathepsins K and S are closely related papain-like cysteine peptidases and potential therapeutic targets for metabolic and inflammatory diseases such as osteoporosis and arthritis. Here we describe the reduction of a previously characterized succinimide (2,5-dioxopyrrolidine)-containing hyperbolic inhibitor of cathepsin K (methyl (RS)-N-[1-(4-methoxyphenyl)-2,5-dioxopyrrolidin-3-yl]glycinate), to obtain a better and more selective compound (compound 4a - methyl (2,5-dioxopyrrolidin-3-yl)glycinate), which acted as a hyperbolic mixed inhibitor/activator similar to already known allosteric effectors of cathepsin K. We then investigated the potential of the succinimide scaffold as inhibitors of cathepsins K and/or S and synthesized a library of such compounds by 1,4-addition of α-amino acid esters and related compounds to N-substituted maleimides. From the generated library, we identified the first small molecule hyperbolic inhibitors of cathepsin S (methyl ((R)-2,5-dioxopyrrolidin-3-yl)-l-threoninate (compound R-4c) and 3-{[(1S,2R,3'S)-2-hydroxycyclohexyl]amino}pyrrolidine-2,5-dione (compound (1S,2R,3'S-10)). The former acted via a similar mechanism to compound 4a, while the latter was a hyperbolic specific inhibitor of cathepsin S. Given the versatility of the scaffold, the identified compounds will be used as the basis for the development of high-affinity hyperbolic inhibitors of the individual peptidases and to explore the potential of hyperbolic inhibitors for the inhibition of cysteine cathepsins in in vitro models.
Collapse
Affiliation(s)
- Tjaša Goričan
- University of Ljubljana, Faculty of Chemistry and Chemical Technology, Department of Chemistry and Biochemistry, Večna pot 113, SI-1000 Ljubljana, Slovenia
| | - Luka Ciber
- University of Ljubljana, Faculty of Chemistry and Chemical Technology, Department of Chemistry and Biochemistry, Večna pot 113, SI-1000 Ljubljana, Slovenia
| | - Nejc Petek
- University of Ljubljana, Faculty of Chemistry and Chemical Technology, Department of Chemistry and Biochemistry, Večna pot 113, SI-1000 Ljubljana, Slovenia
| | - Jurij Svete
- University of Ljubljana, Faculty of Chemistry and Chemical Technology, Department of Chemistry and Biochemistry, Večna pot 113, SI-1000 Ljubljana, Slovenia
| | - Marko Novinec
- University of Ljubljana, Faculty of Chemistry and Chemical Technology, Department of Chemistry and Biochemistry, Večna pot 113, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
36
|
Holyavka M, Faizullin D, Koroleva V, Olshannikova S, Zakhartchenko N, Zuev Y, Kondratyev M, Zakharova E, Artyukhov V. Novel biotechnological formulations of cysteine proteases, immobilized on chitosan. Structure, stability and activity. Int J Biol Macromol 2021; 180:161-176. [PMID: 33676977 DOI: 10.1016/j.ijbiomac.2021.03.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 02/20/2021] [Accepted: 03/03/2021] [Indexed: 12/14/2022]
Abstract
Bromelain, papain, and ficin are studied the most for meat tenderization, but have limited application due to their short lifetime. The aim of this work is to identify the adsorption mechanisms of these cysteine proteases on chitosan to improve the enzymes' stability. It is known that immobilization can lead to a significant loss of enzyme activity, which we observed during the sorption of bromelain (protease activity compared to soluble enzyme is 49% for medium and 64% for high molecular weight chitosan), papain (34 and 28% respectively) and ficin (69 and 70% respectively). Immobilization on the chitosan matrix leads to a partial destruction of protein helical structure (from 5 to 19%). Using computer modelling, we have shown that the sorption of cysteine proteases on chitosan is carried out by molecule regions located on the border of domains L and R, including active cites of the enzymes, which explains the decrease in their catalytic activity upon immobilization. The immobilization on chitosan does not shift the optimal range of pH (7.5) and temperature values (60 °C for bromelain and papain, 37-60 °C for ficin), but significantly increases the stability of biocatalysts (from 5.8 times for bromelain to 7.6 times for papain).
Collapse
Affiliation(s)
- Marina Holyavka
- Voronezh State University, Universitetskaya sq. 1, Voronezh 394018, Russian Federation; Sevastopol State University, Universitetskaya st. 33, Sevastopol 299053, Russian Federation.
| | - Dzhigangir Faizullin
- Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center of RAS, 2/31, Lobachevsky st., Kazan 420111, Russian Federation
| | - Victoria Koroleva
- Voronezh State University, Universitetskaya sq. 1, Voronezh 394018, Russian Federation
| | - Svetlana Olshannikova
- Voronezh State University, Universitetskaya sq. 1, Voronezh 394018, Russian Federation
| | - Nataliya Zakhartchenko
- Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center of RAS, 2/31, Lobachevsky st., Kazan 420111, Russian Federation
| | - Yuriy Zuev
- Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center of RAS, 2/31, Lobachevsky st., Kazan 420111, Russian Federation
| | - Maxim Kondratyev
- Institute of Cell Biophysics of the Russian Academy of Sciences, Institutskaya st. 3, Puschino, Moscow region 142290, Russian Federation
| | - Ekaterina Zakharova
- Institute of Cell Biophysics of the Russian Academy of Sciences, Institutskaya st. 3, Puschino, Moscow region 142290, Russian Federation
| | - Valeriy Artyukhov
- Voronezh State University, Universitetskaya sq. 1, Voronezh 394018, Russian Federation
| |
Collapse
|
37
|
Sun YX, Chen C, Xu WJ, Abbas MN, Mu FF, Ding WJ, Zhang HJ, Li J. Functions of Bombyx mori cathepsin L-like in innate immune response and anti-microbial autophagy. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 116:103927. [PMID: 33197480 DOI: 10.1016/j.dci.2020.103927] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/07/2020] [Accepted: 11/10/2020] [Indexed: 06/11/2023]
Abstract
Cathepsins belongs to the cysteine protease family, which are activated by an acidic environment. They play essential biological roles in the innate immunity and development of animals. Here, we identified a 62 kDa cathepsin L-like protease from the silkworm Bombyx mori. It contained putative conserved domains, including an I29 inhibitor domain and a peptidase C1A domain. The expression analysis revealed that cathepsin L-like was highly produced in the fat body, and 20-hydroxyecdysone (20 E) induced its expression. After challenge with three different types of heat-killed pathogens (Escherichia coli, Beauveria bassiana, and Bacillus cereus), the mRNA levels of cathepsin L-like significantly increased and displayed variable expression patterns in the immune tissues, suggesting its potential role in the innate immune response. The suppression of cathepsin L-like altered the expression of immune-related genes associated with the Toll and IMD pathway. Besides, autophagy-related genes such as Atg6, Atg8, VAMP2, Vps4, and syntaxin expression were also altered, indicating that cathepsin L-like regulates innate immunity and autophagy. Fluorescence microscopic analysis exhibited that cathepsin L-like was localized in the cytoplasm, and it was activated and dispersed throughout the cytoplasm and nucleus following the induction of anti-microbial autophagy. Altogether, our data suggest that cathepsin L-like may regulate the innate immune response and anti-microbial autophagy in the silkworm, B. mori.
Collapse
Affiliation(s)
- Yu-Xuan Sun
- College of Life Sciences, Huaibei Normal University, Huaibei, Anhui, China
| | - Chen Chen
- College of Life Sciences, Huaibei Normal University, Huaibei, Anhui, China
| | - Wen-Jie Xu
- College of Life Sciences, Huaibei Normal University, Huaibei, Anhui, China
| | - Muhammad Nadeem Abbas
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Fang-Fang Mu
- College of Life Sciences, Huaibei Normal University, Huaibei, Anhui, China
| | - Wen-Jing Ding
- College of Life Sciences, Huaibei Normal University, Huaibei, Anhui, China
| | - Hai-Jun Zhang
- College of Life Sciences, Huaibei Normal University, Huaibei, Anhui, China.
| | - Jun Li
- College of Life Sciences, Huaibei Normal University, Huaibei, Anhui, China.
| |
Collapse
|
38
|
Ulčakar L, Novinec M. Inhibition of Human Cathepsins B and L by Caffeic Acid and Its Derivatives. Biomolecules 2020; 11:E31. [PMID: 33383850 PMCID: PMC7824550 DOI: 10.3390/biom11010031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/18/2020] [Accepted: 12/25/2020] [Indexed: 12/15/2022] Open
Abstract
Caffeic acid (CA) and its derivatives caffeic acid phenethyl ester (CAPE) and chlorogenic acid (CGA) are phenolic compounds of plant origin with a wide range of biological activities. Here, we identify and characterize their inhibitory properties against human cathepsins B and L, potent, ubiquitously expressed cysteine peptidases involved in protein turnover and homeostasis, as well as pathological conditions, such as cancer. We show that CAPE and CGA inhibit both peptidases, while CA shows a preference for cathepsin B, resulting in the strongest inhibition among these combinations. All compounds are linear (complete) inhibitors acting via mixed or catalytic mechanisms. Cathepsin B is more strongly inhibited at pH 7.4 than at 5.5, and CA inhibits its endopeptidase activity preferentially over its peptidyl-dipeptidase activity. Altogether, the results identify the CA scaffold as a promising candidate for the development of cathepsin B inhibitors, specifically targeting its endopeptidase activity associated with pathological proteolysis of extracellular substrates.
Collapse
Affiliation(s)
| | - Marko Novinec
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, 1000 Ljubljana, Slovenia;
| |
Collapse
|
39
|
Denessiouk K, Uversky VN, Permyakov SE, Permyakov EA, Johnson MS, Denesyuk AI. Papain-like cysteine proteinase zone (PCP-zone) and PCP structural catalytic core (PCP-SCC) of enzymes with cysteine proteinase fold. Int J Biol Macromol 2020; 165:1438-1446. [PMID: 33058970 PMCID: PMC7548629 DOI: 10.1016/j.ijbiomac.2020.10.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 09/13/2020] [Accepted: 10/03/2020] [Indexed: 12/01/2022]
Abstract
There are several families of cysteine proteinases with different folds – for example the (chymo)trypsin fold family and papain-like fold family – but in both families the hydrolase activity of cysteine proteinases requires a cysteine residue as the catalytic nucleophile. In this work, we have analyzed the topology of the active site regions in 146 three-dimensional structures of proteins belonging to the Papain-like Cysteine Proteinase (PCP) superfamily, which includes papain as a typical representative of this protein superfamily. All analyzed enzymes contain a unique structurally closed conformation – a “PCP-Zone” – which can be divided into two groups, Class A and Class B. Eight structurally conserved amino acids of the PCP-Zone form a common Structural Core. The Structural Core, catalytic nucleophile, catalytic base and residue Xaa – which stabilizes the side-chain conformation of the catalytic base – make up a PCP Structural Catalytic Core (PCP-SCC). The PCP-SCC of Class A and Class B are divided into 5 and 2 types, respectively. Seven variants of the mutual arrangement of the amino-acid side chains of the catalytic triad – nucleophile, base and residue Xaa – within the same fold clearly demonstrate how enzymes with the papain-like fold adapt to the need to perform diverse functions in spite of their limited structural diversity. The roles of both the PCP-Zone of SARS-CoV-2-PLpro described in this study and the NBCZone of SARS-CoV-2-3CLpro presented in our earlier article (Denesyuk AI, Johnson MS, Salo-Ahen OMH, Uversky VN, Denessiouk K. Int J Biol Macromol. 2020;153:399-411) that are in contacts with inhibitors are discussed.
Collapse
Affiliation(s)
- Konstantin Denessiouk
- Structural Bioinformatics Laboratory, Faculty of Science and Engineering, Biochemistry, Åbo Akademi University, Turku 20520, Finland
| | - Vladimir N Uversky
- Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino 142290, Russia; Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| | - Sergei E Permyakov
- Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino 142290, Russia
| | - Eugene A Permyakov
- Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino 142290, Russia
| | - Mark S Johnson
- Structural Bioinformatics Laboratory, Faculty of Science and Engineering, Biochemistry, Åbo Akademi University, Turku 20520, Finland
| | - Alexander I Denesyuk
- Structural Bioinformatics Laboratory, Faculty of Science and Engineering, Biochemistry, Åbo Akademi University, Turku 20520, Finland; Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino 142290, Russia
| |
Collapse
|
40
|
Soares AM, Gonçalves LM, Ferreira RD, de Souza JM, Fangueiro R, Alves MM, Carvalho FA, Mendes AN, Cantanhêde W. Immobilization of papain enzyme on a hybrid support containing zinc oxide nanoparticles and chitosan for clinical applications. Carbohydr Polym 2020; 243:116498. [DOI: 10.1016/j.carbpol.2020.116498] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 04/20/2020] [Accepted: 05/20/2020] [Indexed: 02/08/2023]
|
41
|
Cruz WT, Bezerra EHS, Ramos MV, Rocha BAM, Medina MC, Demarco D, Carvalho CPS, Oliveira JS, Sousa JS, Souza PFN, Freire VN, da Silva FMS, Freitas CDT. Crystal structure and specific location of a germin-like protein with proteolytic activity from Thevetia peruviana. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2020; 298:110590. [PMID: 32771148 DOI: 10.1016/j.plantsci.2020.110590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/26/2020] [Accepted: 07/01/2020] [Indexed: 06/11/2023]
Abstract
Peruvianin-I is a cysteine peptidase (EC 3.4.22) purified from Thevetia peruviana. Previous studies have shown that it is the only germin-like protein (GLP) with proteolytic activity described so far. In this work, the X-ray crystal structure of peruvianin-I was determined to a resolution of 2.15 Å (PDB accession number: 6ORM) and its specific location was evaluated by different assays. Its overall structure shows an arrangement composed of a homohexamer (a trimer of dimers) where each monomer exhibits a typical β-barrel fold and two glycosylation sites (Asn55 and Asn144). Analysis of its active site confirmed the absence of essential amino acids for typical oxalate oxidase activity of GLPs. Details of the active site and molecular docking results, using a specific cysteine peptidase inhibitor (iodoacetamide), were used to discuss a plausible mechanism for proteolytic activity of peruvianin-I. Histological analyses showed that T. peruviana has articulated anastomosing laticifers, i.e., rows of cells which merge to form continuous tubes throughout its green organs. Moreover, peruvianin-I was detected exclusively in the latex. Because latex peptidases have been described as defensive molecules against insects, we hypothesize that peruvianin-I contributes to protect T. peruviana plants against herbivory.
Collapse
Affiliation(s)
- Wallace T Cruz
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Ceará, Campus do Pici, CEP 60.440-554, Fortaleza, Ceará, Brazil
| | - Eduardo H S Bezerra
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Ceará, Campus do Pici, CEP 60.440-554, Fortaleza, Ceará, Brazil
| | - Márcio V Ramos
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Ceará, Campus do Pici, CEP 60.440-554, Fortaleza, Ceará, Brazil
| | - Bruno A M Rocha
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Ceará, Campus do Pici, CEP 60.440-554, Fortaleza, Ceará, Brazil
| | - Maria C Medina
- Departamento de Botânica, Instituto de Biociências, Universidade de São Paulo, CEP 05.508-090, São Paulo, São Paulo, Brazil
| | - Diego Demarco
- Departamento de Botânica, Instituto de Biociências, Universidade de São Paulo, CEP 05.508-090, São Paulo, São Paulo, Brazil
| | - Cristina Paiva S Carvalho
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Ceará, Campus do Pici, CEP 60.440-554, Fortaleza, Ceará, Brazil
| | - Jefferson S Oliveira
- Departamento de Biomedicina, Universidade Federal do Delta do Parnaíba, Campus Ministro Reis Velloso, Parnaíba, Piauí, Brazil
| | - Jeanlex S Sousa
- Departamento de Física, Universidade Federal de Ceará, Fortaleza, Brazil
| | - Pedro F N Souza
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Ceará, Campus do Pici, CEP 60.440-554, Fortaleza, Ceará, Brazil
| | - Valder N Freire
- Departamento de Física, Universidade Federal de Ceará, Fortaleza, Brazil
| | | | - Cleverson D T Freitas
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Ceará, Campus do Pici, CEP 60.440-554, Fortaleza, Ceará, Brazil.
| |
Collapse
|
42
|
Sparks ME, Bansal R, Benoit JB, Blackburn MB, Chao H, Chen M, Cheng S, Childers C, Dinh H, Doddapaneni HV, Dugan S, Elpidina EN, Farrow DW, Friedrich M, Gibbs RA, Hall B, Han Y, Hardy RW, Holmes CJ, Hughes DST, Ioannidis P, Cheatle Jarvela AM, Johnston JS, Jones JW, Kronmiller BA, Kung F, Lee SL, Martynov AG, Masterson P, Maumus F, Munoz-Torres M, Murali SC, Murphy TD, Muzny DM, Nelson DR, Oppert B, Panfilio KA, Paula DP, Pick L, Poelchau MF, Qu J, Reding K, Rhoades JH, Rhodes A, Richards S, Richter R, Robertson HM, Rosendale AJ, Tu ZJ, Velamuri AS, Waterhouse RM, Weirauch MT, Wells JT, Werren JH, Worley KC, Zdobnov EM, Gundersen-Rindal DE. Brown marmorated stink bug, Halyomorpha halys (Stål), genome: putative underpinnings of polyphagy, insecticide resistance potential and biology of a top worldwide pest. BMC Genomics 2020; 21:227. [PMID: 32171258 PMCID: PMC7071726 DOI: 10.1186/s12864-020-6510-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 01/20/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Halyomorpha halys (Stål), the brown marmorated stink bug, is a highly invasive insect species due in part to its exceptionally high levels of polyphagy. This species is also a nuisance due to overwintering in human-made structures. It has caused significant agricultural losses in recent years along the Atlantic seaboard of North America and in continental Europe. Genomic resources will assist with determining the molecular basis for this species' feeding and habitat traits, defining potential targets for pest management strategies. RESULTS Analysis of the 1.15-Gb draft genome assembly has identified a wide variety of genetic elements underpinning the biological characteristics of this formidable pest species, encompassing the roles of sensory functions, digestion, immunity, detoxification and development, all of which likely support H. halys' capacity for invasiveness. Many of the genes identified herein have potential for biomolecular pesticide applications. CONCLUSIONS Availability of the H. halys genome sequence will be useful for the development of environmentally friendly biomolecular pesticides to be applied in concert with more traditional, synthetic chemical-based controls.
Collapse
Affiliation(s)
- Michael E Sparks
- USDA-ARS Invasive Insect Biocontrol and Behavior Laboratory, Beltsville, MD, 20705, USA.
| | - Raman Bansal
- USDA-ARS San Joaquin Valley Agricultural Sciences Center, Parlier, CA, 93648, USA
| | - Joshua B Benoit
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH, 45221, USA
| | - Michael B Blackburn
- USDA-ARS Invasive Insect Biocontrol and Behavior Laboratory, Beltsville, MD, 20705, USA
| | - Hsu Chao
- Department of Human and Molecular Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Mengyao Chen
- Department of Entomology, University of Maryland, College Park, MD, 20742, USA
| | - Sammy Cheng
- Department of Biology, University of Rochester, Rochester, NY, 14627, USA
| | | | - Huyen Dinh
- Department of Human and Molecular Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Harsha Vardhan Doddapaneni
- Department of Human and Molecular Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Shannon Dugan
- Department of Human and Molecular Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Elena N Elpidina
- A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, 119911, Russia
| | - David W Farrow
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH, 45221, USA
| | - Markus Friedrich
- Department of Biological Sciences, Wayne State University, Detroit, MI, 48201, USA
| | - Richard A Gibbs
- Department of Human and Molecular Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Brantley Hall
- Department of Biochemistry, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Yi Han
- Department of Human and Molecular Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Richard W Hardy
- Department of Biology, Indiana University, Bloomington, IN, 47405, USA
| | - Christopher J Holmes
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH, 45221, USA
| | - Daniel S T Hughes
- Department of Human and Molecular Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Panagiotis Ioannidis
- Department of Genetic Medicine and Development, University of Geneva Medical School and Swiss Institute of Bioinformatics, 1211, Geneva, Switzerland
- Present address: Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, 73100, Heraklion, Crete, Greece
| | | | - J Spencer Johnston
- Department of Entomology, Texas A&M University, College Station, TX, 77843, USA
| | - Jeffery W Jones
- Department of Biological Sciences, Wayne State University, Detroit, MI, 48201, USA
| | - Brent A Kronmiller
- Center for Genome Research and Biocomputing, Oregon State University, Corvallis, OR, 97331, USA
| | - Faith Kung
- Department of Entomology, University of Maryland, College Park, MD, 20742, USA
| | - Sandra L Lee
- Department of Human and Molecular Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Alexander G Martynov
- Center for Data-Intensive Biomedicine and Biotechnology, Skolkovo Institute of Science and Technology, Skolkovo, 143025, Russia
| | - Patrick Masterson
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, 20894, USA
| | - Florian Maumus
- URGI, INRA, Université Paris-Saclay, 78026, Versailles, France
| | - Monica Munoz-Torres
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Shwetha C Murali
- Department of Human and Molecular Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Terence D Murphy
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, 20894, USA
| | - Donna M Muzny
- Department of Human and Molecular Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - David R Nelson
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Brenda Oppert
- USDA-ARS Center for Grain and Animal Health Research, Manhattan, KS, 66502, USA
| | - Kristen A Panfilio
- Developmental Biology, Institute for Zoology: University of Cologne, 50674, Cologne, Germany
- School of Life Sciences, University of Warwick, Gibbet Hill Campus, Coventry, CV4 7AL, United Kingdom
| | - Débora Pires Paula
- EMBRAPA Genetic Resources and Biotechnology, Brasília, DF, 70770-901, Brazil
| | - Leslie Pick
- Department of Entomology, University of Maryland, College Park, MD, 20742, USA
| | | | - Jiaxin Qu
- Department of Human and Molecular Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Katie Reding
- Department of Entomology, University of Maryland, College Park, MD, 20742, USA
| | - Joshua H Rhoades
- USDA-ARS Invasive Insect Biocontrol and Behavior Laboratory, Beltsville, MD, 20705, USA
| | - Adelaide Rhodes
- Larner College of Medicine, The University of Vermont, Burlington, VT, 05452, USA
| | - Stephen Richards
- Department of Human and Molecular Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
- Present address: Earth BioGenome Project, University of California, Davis, Davis, CA, 95616, USA
| | - Rose Richter
- Department of Biology, University of Rochester, Rochester, NY, 14627, USA
| | - Hugh M Robertson
- Department of Entomology, University of Illinois, Urbana-Champaign, IL, 61801, USA
| | - Andrew J Rosendale
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH, 45221, USA
| | - Zhijian Jake Tu
- Department of Biochemistry, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Arun S Velamuri
- USDA-ARS Invasive Insect Biocontrol and Behavior Laboratory, Beltsville, MD, 20705, USA
| | - Robert M Waterhouse
- Department of Ecology and Evolution, University of Lausanne and Swiss Institute of Bioinformatics, 1015, Lausanne, Switzerland
| | - Matthew T Weirauch
- Division of Biomedical Informatics, and Division of Developmental Biology, Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Jackson T Wells
- Center for Genome Research and Biocomputing, Oregon State University, Corvallis, OR, 97331, USA
| | - John H Werren
- Department of Biology, University of Rochester, Rochester, NY, 14627, USA
| | - Kim C Worley
- Department of Human and Molecular Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Evgeny M Zdobnov
- Department of Genetic Medicine and Development, University of Geneva Medical School and Swiss Institute of Bioinformatics, 1211, Geneva, Switzerland
| | | |
Collapse
|
43
|
Gimenes NC, Silveira E, Tambourgi EB. An Overview of Proteases: Production, Downstream Processes and Industrial Applications. SEPARATION & PURIFICATION REVIEWS 2019. [DOI: 10.1080/15422119.2019.1677249] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
| | - Edgar Silveira
- Biotechnology Institute, Federal University of Uberlandia, Uberlandia, Minas Gerais, Brazil
- Brazilian Savanna’s, Diversity Research Center, Federal University of Uberlandia, Uberlandia, Minas Gerais, Brazil
| | | |
Collapse
|
44
|
Rebernik M, Snoj T, Klemenčič M, Novinec M. Interplay between tetrameric structure, enzymatic activity and allosteric regulation of human dipeptidyl-peptidase I. Arch Biochem Biophys 2019; 675:108121. [DOI: 10.1016/j.abb.2019.108121] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/10/2019] [Accepted: 09/27/2019] [Indexed: 02/06/2023]
|
45
|
Processing and Maturation of Cathepsin C Zymogen: A Biochemical and Molecular Modeling Analysis. Int J Mol Sci 2019; 20:ijms20194747. [PMID: 31557781 PMCID: PMC6801622 DOI: 10.3390/ijms20194747] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 09/11/2019] [Accepted: 09/18/2019] [Indexed: 11/16/2022] Open
Abstract
Cysteine cathepsin C (CatC) is a ubiquitously expressed, lysosomal aminopeptidase involved in the activation of zymogens of immune-cell-associated serine proteinases (elastase, cathepsin G, proteinase 3, neutrophil serine proteinase 4, lymphocyte granzymes, and mast cell chymases). CatC is first synthetized as an inactive zymogen containing an intramolecular chain propeptide, the dimeric form of which is processed into the mature tetrameric form by proteolytic cleavages. A molecular modeling analysis of proCatC indicated that its propeptide displayed a similar fold to those of other lysosomal cysteine cathepsins, and could be involved in dimer formation. Our in vitro experiments revealed that human proCatC was processed and activated by CatF, CatK, and CatV in two consecutive steps of maturation, as reported for CatL and CatS previously. The unique positioning of the propeptide domains in the proCatC dimer complex allows this order of cleavages to be understood. The missense mutation Leu172Pro within the propeptide region associated with the Papillon-Lefèvre and Haim-Munk syndrome altered the proform stability as well as the maturation of the recombinant Leu172Pro proform.
Collapse
|
46
|
Jakoš T, Pišlar A, Jewett A, Kos J. Cysteine Cathepsins in Tumor-Associated Immune Cells. Front Immunol 2019; 10:2037. [PMID: 31555270 PMCID: PMC6724555 DOI: 10.3389/fimmu.2019.02037] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/12/2019] [Indexed: 12/23/2022] Open
Abstract
Cysteine cathepsins are key regulators of the innate and adaptive arms of the immune system. Their expression, activity, and subcellular localization are associated with the distinct development and differentiation stages of immune cells. They promote the activation of innate myeloid immune cells since they contribute to toll-like receptor signaling and to cytokine secretion. Furthermore, they control lysosomal biogenesis and autophagic flux, thus affecting innate immune cell survival and polarization. They also regulate bidirectional communication between the cell exterior and the cytoskeleton, thus influencing cell interactions, morphology, and motility. Importantly, cysteine cathepsins contribute to the priming of adaptive immune cells by controlling antigen presentation and are involved in cytotoxic granule mediated killing in cytotoxic T lymphocytes and natural killer cells. Cathepins'aberrant activity can be prevented by their endogenous inhibitors, cystatins. However, dysregulated proteolysis contributes significantly to tumor progression also by modulation of the antitumor immune response. Especially tumor-associated myeloid cells, such as tumor-associated macrophages and myeloid-derived suppressor cells, which are known for their tumor promoting and immunosuppressive functions, constitute the major source of excessive cysteine cathepsin activity in cancer. Since they are enriched in the tumor microenvironment, cysteine cathepsins represent exciting targets for development of new diagnostic and therapeutic moieties.
Collapse
Affiliation(s)
- Tanja Jakoš
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Anja Pišlar
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Anahid Jewett
- UCLA School of Dentistry and Medicine, Los Angeles, CA, United States
| | - Janko Kos
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia.,Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia
| |
Collapse
|
47
|
Ji C, Liang Y, Ge F, Yang L, Wang Q. Inhibitory Covalent Labeling and Clickable-Eu-Tagging-Based ICPMS: Measurement of pH-Dependent Absolute Activities of the Cathepsins in Hepatocyte Lysosomes. Anal Chem 2019; 91:7032-7038. [PMID: 31072096 DOI: 10.1021/acs.analchem.9b01662] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We report an inhibitory covalent labeling and clickable-element-tagging strategy for measuring the absolute activity of a protease in cells using inductively coupled plasma mass spectrometry (ICPMS). Epoxysuccinyl-leucine-tyrosine-6-aminocaproic-lysine-amino-Boc-alkyne (epoxysuccinyl-LYK-alkyne) was designed and synthesized to achieve irreversibly labeling of the cysteine cathepsins, recording their momentary activities. L and Y assisted epoxysuccinyl-LYK-alkyne in accessing the deprotonated -S- of Cys25, located at the bottom of the long cathepsin active domain. Quantitative Eu-tagging was followed using azido-DOTA-Eu through a bioorthogonal 1:1 copper-catalyzed azide-alkyne-cycloaddition click reaction. The Eu tag could be absolutely quantified using 153Eu-species-nonspecific-isotope-dilution ICPMS coupled with HPLC, serving as a Eu ruler and allowing us to simultaneously measure the pH-dependent activities of cathepsins B, L, and S as well as the pH in the lysosomal microenvironment of liver cancerous C7721 and paracancerous C7701 cells. As long as suitable labeling molecules and elemental tags are designed and synthesized, we believe that such a tandem labeling and tagging ICPMS approach can be applied to the measurement of the activities of other proteases in cells, providing more accurate information on the proteases' biofunctions and thus implementing precise clinical diagnoses.
Collapse
Affiliation(s)
- Caixia Ji
- Department of Chemistry & the MOE Key Lab of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering , Xiamen University , Xiamen 361005 , China
| | - Yong Liang
- Department of Chemistry & the MOE Key Lab of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering , Xiamen University , Xiamen 361005 , China
| | - Fuchun Ge
- Department of Chemistry & the MOE Key Lab of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering , Xiamen University , Xiamen 361005 , China
| | - Limin Yang
- Department of Chemistry & the MOE Key Lab of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering , Xiamen University , Xiamen 361005 , China
| | - Qiuquan Wang
- Department of Chemistry & the MOE Key Lab of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering , Xiamen University , Xiamen 361005 , China.,State Key Lab of Marine Environmental Science , Xiamen University , Xiamen 361005 , China
| |
Collapse
|
48
|
Xavier MA, Tirloni L, Torquato R, Tanaka A, Pinto AFM, Diedrich JK, Yates JR, da Silva Vaz I, Seixas A, Termignoni C. Blood anticlotting activity of a Rhipicephalus microplus cathepsin L-like enzyme. Biochimie 2019; 163:12-20. [PMID: 31059753 DOI: 10.1016/j.biochi.2019.04.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 04/29/2019] [Indexed: 01/05/2023]
Abstract
In parasites, cathepsins are implicated in mechanisms related to organism surveillance and host evasion. Some parasite cathepsins have fibrinogenolytic and fibrinolytic activity, suggesting that they may contribute to maintain blood meal fluidity for extended feeding periods. Here, it is shown that BmGTI (Rhipicephalus [Boophilus] microplus Gut Thrombin Inhibitor), a protein previously described as an inhibitor of fibrinogen hydrolysis and platelet aggregation by thrombin, and BmCL1 (Rhipicephalus [Boophilus] microplus Cathepsin-L like 1) are the same protein, hereinafter referred to using the earliest name (BmCL1). To further characterize BmCL1, Rhipicephalus microplus native and recombinant (rBmCL1) proteins were obtained. Native BmCL1 was isolated using thrombin-affinity chromatography, and it displays thrombin inhibition activity. We subsequently investigated rBmCL1 interaction with thrombin. We show that rBmCL1 and thrombin have a dissociation constant (ΚD) of 130.2 ± 11.2 nM, and this interaction likely occurs due to a more electronegative surface of BmCL1 at pH 7.5 than at pH 5.0, which may favor an electrostatic binding to positively charged thrombin exosites. During BmCL1-thrombin interaction, thrombin is not degraded or inhibited. rBmCL1 impairs thrombin-induced fibrinogen clotting via a fibrinogenolytic activity. Fibrinogen degradation by BmCL1 occurs by the hydrolysis of Aα- and Bβ-chains, generating products similar to those produced by fibrinogenolytic cathepsins from other organisms. In conclusion, BmCL1 likely has an additional role in R. microplus blood digestion, besides its role in hemoglobin degradation at acid pH. BmCL1 fibrinogenolytic activity indicates a proteolytic activity in the neutral lumen of tick midgut, contributing to maintain the fluidity of the ingested blood, which remains to be confirmed in vivo.
Collapse
Affiliation(s)
- Marina Amaral Xavier
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Lucas Tirloni
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, USA
| | - Ricardo Torquato
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Aparecida Tanaka
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Antônio F M Pinto
- Clayton Foundation Peptide Biology Lab, Salk Institute for Biological Studies, USA
| | - Jolene K Diedrich
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, USA
| | - John R Yates
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Itabajara da Silva Vaz
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Adriana Seixas
- Departamento de Farmacociências, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| | - Carlos Termignoni
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
49
|
Mann KS, Sanfaçon H. Expanding Repertoire of Plant Positive-Strand RNA Virus Proteases. Viruses 2019; 11:v11010066. [PMID: 30650571 PMCID: PMC6357015 DOI: 10.3390/v11010066] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/11/2019] [Accepted: 01/12/2019] [Indexed: 12/13/2022] Open
Abstract
Many plant viruses express their proteins through a polyprotein strategy, requiring the acquisition of protease domains to regulate the release of functional mature proteins and/or intermediate polyproteins. Positive-strand RNA viruses constitute the vast majority of plant viruses and they are diverse in their genomic organization and protein expression strategies. Until recently, proteases encoded by positive-strand RNA viruses were described as belonging to two categories: (1) chymotrypsin-like cysteine and serine proteases and (2) papain-like cysteine protease. However, the functional characterization of plant virus cysteine and serine proteases has highlighted their diversity in terms of biological activities, cleavage site specificities, regulatory mechanisms, and three-dimensional structures. The recent discovery of a plant picorna-like virus glutamic protease with possible structural similarities with fungal and bacterial glutamic proteases also revealed new unexpected sources of protease domains. We discuss the variety of plant positive-strand RNA virus protease domains. We also highlight possible evolution scenarios of these viral proteases, including evidence for the exchange of protease domains amongst unrelated viruses.
Collapse
Affiliation(s)
- Krin S Mann
- Summerland Research and Development Centre, Agriculture and Agri-Food Canada, Summerland, BC V0H 1Z0, Canada.
| | - Hélène Sanfaçon
- Summerland Research and Development Centre, Agriculture and Agri-Food Canada, Summerland, BC V0H 1Z0, Canada.
| |
Collapse
|
50
|
Zhang S, Xu Z, Sun H, Sun L, Shaban M, Yang X, Zhu L. Genome-Wide Identification of Papain-Like Cysteine Proteases in Gossypium hirsutum and Functional Characterization in Response to Verticillium dahliae. FRONTIERS IN PLANT SCIENCE 2019; 10:134. [PMID: 30842780 PMCID: PMC6391353 DOI: 10.3389/fpls.2019.00134] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 01/28/2019] [Indexed: 05/12/2023]
Abstract
Cotton, a natural fiber producing crop of huge importance, is often prone to attack of Verticillium dahliae. Papain-like cysteine proteases (PLCPs) constitute a large family in plants and were proposed to involve in plant defense against pathogen attack in a number of studies. However, there is no detailed characterization of PLCP genes in cotton against infection of V. dahliae. In this study, we carried out a genome-wide analysis in cotton and identified seventy-eight PLCPs, which were divided into nine subfamilies based on their evolution phylogeny: RD21 (responsive to desiccation 21), CEP (cysteine endopeptidase), XCP (xylem cysteine peptidase), XBCP3 (xylem bark cysteine peptidase 3), THI, SAG12 (senescence-associated gene 12), RD19 (responsive to desiccation 19), ALP (aleurain-like protease) and CTB (cathepsin B-like). Genes in each subfamily exhibit a similar structure and motif composition. The expression patterns of these genes in different organs were examined, and subfamily RD21 was the most abundant in these families. Expression profiles under abiotic stress showed that thirty-five PLCP genes were induced by multiple stresses. Further transcriptome analysis showed that sixteen PLCP genes were up-regulated in response to V. dahliae in cotton. Among those, GhRD21-7 showed a higher transcription level than most other PLCP genes. Additionally, over-expression of GhRD21-7 led to enhanced resistance and RNAi lines were more susceptible to V. dahliae in cotton. Our results provide valuable information for future functional genomic studies of PLCP gene family in cotton.
Collapse
|