1
|
Rabea EY, Mahmoud ED, Mohamed NK, Ansary ER, Alrouby MR, Shehata RR, Mokhtar YY, Arullampalam P, Hegazy AM, Al-Sabi A, Abd El-Aziz TM. Potential of Venom-Derived Compounds for the Development of New Antimicrobial Agents. Toxins (Basel) 2025; 17:238. [PMID: 40423321 DOI: 10.3390/toxins17050238] [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: 03/16/2025] [Revised: 05/02/2025] [Accepted: 05/09/2025] [Indexed: 05/28/2025] Open
Abstract
The emergence of antimicrobial resistance is a significant challenge in global healthcare, necessitating innovative techniques to address multidrug-resistant pathogens. Multidrug-resistant pathogens like Klebsiella pneumoniae, Acinetobacter baumannii, and Pseudomonas aeruginosa pose significant public health threats, as they are increasingly resistant to common antibiotics, leading to more severe and difficult-to-treat infections. These pathogens are part of the ESKAPE group, which includes Enterococcus faecium, Staphylococcus aureus, and Enterobacter species. Animal venoms, derived from a wide range of species such as snakes, scorpions, spiders, bees, wasps, and ants, represent a rich source of bioactive peptides. Venoms have been a valuable source for drug discovery, providing unique compounds with therapeutic potential. Venom-derived drugs are known for their increased bioactivity, specificity, and stability compared to synthetic alternatives. These compounds are being investigated for various conditions, including treatments for diabetes, pain relief, cancer, and infections, showcasing their remarkable antimicrobial efficacy. In this review, we provide a comprehensive investigation into the potential of venom-derived compounds for developing new antimicrobial agents, including antibacterial, antifungal, antiviral, and antiparasitic therapeutics. Key venom components, including melittin from bee venom, phospholipase A2 from snake venom, and chlorotoxin from scorpion venom, exhibit potent antimicrobial effects through mechanisms such as membrane disruption, enzymatic inhibition, and immune modulation. We also explore the challenges related to the development and clinical use of venom-derived antimicrobials, including toxicity, stability, and delivery mechanisms. These compounds hold immense promise as transformative tools against resistant pathogens, offering a unique avenue for groundbreaking advancements in antimicrobial research and therapeutic development.
Collapse
Affiliation(s)
- Esraa Yasser Rabea
- Biochemistry Division, Chemistry Department, Faculty of Science, Minia University, El-Minia 61519, Egypt
| | - Esraa Dakrory Mahmoud
- Biochemistry Division, Chemistry Department, Faculty of Science, Minia University, El-Minia 61519, Egypt
| | - Nada Khaled Mohamed
- Biochemistry Division, Chemistry Department, Faculty of Science, Minia University, El-Minia 61519, Egypt
| | - Erada Rabea Ansary
- Biochemistry Division, Chemistry Department, Faculty of Science, Minia University, El-Minia 61519, Egypt
| | - Mahmoud Roushdy Alrouby
- Biochemistry Division, Chemistry Department, Faculty of Science, Minia University, El-Minia 61519, Egypt
| | - Rabab Reda Shehata
- Biochemistry Division, Chemistry Department, Faculty of Science, Minia University, El-Minia 61519, Egypt
| | - Youssef Yasser Mokhtar
- Biochemistry Division, Chemistry Department, Faculty of Science, Minia University, El-Minia 61519, Egypt
| | - Prakash Arullampalam
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ahmed M Hegazy
- Zoology Department, Faculty of Science, Minia University, El-Minia 61519, Egypt
| | - Ahmed Al-Sabi
- College of Integrative Studies, Abdullah Al Salem University, Khaldiya 72303, Kuwait
| | - Tarek Mohamed Abd El-Aziz
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO 63110, USA
- Zoology Department, Faculty of Science, Minia University, El-Minia 61519, Egypt
| |
Collapse
|
2
|
Castro-Amorim J, Pinto AV, Mukherjee AK, Ramos MJ, Fernandes PA. Beyond Fang's fury: a computational study of the enzyme-membrane interaction and catalytic pathway of the snake venom phospholipase A 2 toxin. Chem Sci 2025; 16:1974-1985. [PMID: 39759936 PMCID: PMC11694569 DOI: 10.1039/d4sc06511e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/19/2024] [Indexed: 01/07/2025] Open
Abstract
Snake venom-secreted phospholipases A2 (svPLA2s) are critical, highly toxic enzymes present in almost all snake venoms. Upon snakebite envenomation, svPLA2s hydrolyze cell membrane phospholipids and induce pathological effects such as paralysis, myonecrosis, inflammation, or pain. Despite its central importance in envenomation, the chemical mechanism of svPLA2s is poorly understood, with detrimental consequences for the design of small-molecule snakebite antidotes, which is highly undesirable given the gravity of the epidemiological data that ranks snakebite as the deadliest neglected tropical disease. We study a member of the svPLA2 family, the Myotoxin-I, which is part of the venom of the Central American pit viper terciopelo (Bothrops asper), a ubiquitous but highly aggressive and dangerous species responsible for the most problematic snakebites in its habitat. Furthermore, PLA2 enzymes are a paradigm of interfacial enzymology, as the complex membrane-enzyme interaction is as important as is crucial for its catalytic process. Here, we explore the detailed interaction between svPLA2 and a 1 : 1 POPC/POPS membrane, and how enzyme binding affects membrane structure and dynamics. We further investigated the two most widely accepted reaction mechanisms for svPLA2s: the 'single-water mechanism' and the 'assisted-water mechanism', using umbrella sampling simulations at the PBE/MM level of theory. We demonstrate that both pathways are catalytically viable. While both pathways occur in two steps, the single-water mechanism yielded a lower activation free energy barrier (20.14 kcal mol-1) for POPC hydrolysis, consistent with experimental and computational values obtained for human PLA2. The reaction mechanisms are similar, albeit not identical, and can be generalized to svPLA2 from most viper species. Furthermore, our findings demonstrate that the sole small molecule inhibitor currently undergoing clinical trials for snakebite is a perfect transition state analog. Thus, understanding snake venom sPLA2 chemistry will help find new, effective small molecule inhibitors with anti-snake venom efficacy.
Collapse
Affiliation(s)
- Juliana Castro-Amorim
- LAQV/Requimte, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto Rua do Campo Alegre, s/n 4169-007 Porto Portugal
| | - Alexandre V Pinto
- LAQV/Requimte, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto Rua do Campo Alegre, s/n 4169-007 Porto Portugal
| | - Ashis K Mukherjee
- Institute of Advanced Study in Science and Technology Vigyan Path Garchuk, Paschim Boragaon Guwahati-781035 Assam India
| | - Maria J Ramos
- LAQV/Requimte, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto Rua do Campo Alegre, s/n 4169-007 Porto Portugal
| | - Pedro A Fernandes
- LAQV/Requimte, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto Rua do Campo Alegre, s/n 4169-007 Porto Portugal
| |
Collapse
|
3
|
Uren RT, Ritchie ME, Wong AW, Ludeman JP, Uno E, Narayana VK, De Souza DP, Sviridov D, Kluck RM. A lipid signature of BAK-driven apoptotic pore formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.16.618570. [PMID: 39463966 PMCID: PMC11507859 DOI: 10.1101/2024.10.16.618570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Apoptotic cell death is regulated by the BCL-2 protein family, with clusters of BAK or BAX homodimers driving pore formation in the mitochondrial outer membrane via a poorly understood process. There is growing evidence that, in addition to BAK and BAX, lipids play an important role in pore formation. Towards a better understanding of the lipidic drivers of apoptotic pore formation in isolated mitochondria, two complementary approaches were taken. Firstly, the lipids released during BAK-mediated pore formation were measured with targeted lipidomics, revealing enrichment of long chain polyunsaturated lysophospholipids (LPLs) in the released fraction. In contrast, the BAK protein was not released suggesting that BAK and LPLs locate to distinct microdomains. Secondly, added cholesterol not only prevented pore formation but prevented the clustering of BAK homodimers. Our data lead us to a model in which BAK clustering triggers formation of a separate microdomain rich in LPLs that can progress to lipid shedding and the opening of a lipid-lined pore. Pore stabilisation and growth may be due to BAK dimers then moving to the pore edge. Our BAK-lipid microdomain model supports the heterogeneity of BAK assemblies, and the observed lipid-release signature gives new insight into the genesis of the apoptotic pore.
Collapse
|
4
|
Bacha AB, Alonazi M. Effective Soybean Oil Degumming by Immobilized Phospholipases A 2 from Walterinnesia aegyptia Venom. ACS OMEGA 2024; 9:21322-21332. [PMID: 38764629 PMCID: PMC11097375 DOI: 10.1021/acsomega.4c01558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 05/21/2024]
Abstract
Enzymatic degumming utilizing phospholipase enzymes could be used in ecologically friendly procedures with enhanced oil recovery yields. In this study, two phospholipases A2 of group I and II, WaPLA2-I and WaPLA2-II, from the snake venom of Saudi Walterinnesia aegyptia were evaluated for soybean oil degumming after being immobilized on three different support materials (calcium alginate (CA), CA-gelatin (CAG), and CA-chitosan (CAC), and cross-linked with glutaraldehyde). Higher yields of CAC-immobilized PLA2-I (85 ± 3%) and PLA2-II (87 ± 3.6%) compared to CAG (77.3 ± 2.1 and 79 ± 2.6%, respectively) and CA beads (55.7 ± 2.5% and 57.3 ± 3.1%, respectively) were observed. In addition, the optimal temperature of immobilized WaPLA2-I and WaPLA2-II increased from 45 to 55 °C and from 55 to 65 °C, respectively. Their stability at high temperatures was also significantly enhanced covering a larger range (70-80 °C). Likewise, the pH/activity profile of WaPLA2 was greatly expanded upon immobilization with the pH-optima being shifted by 0.5 to 1 pH unit to the basic side. Similarly, the stability of WaPLA2s in the presence of organic solvents was also significantly improved, while the affinity for calcium and bile salt was the same for both free and immobilized enzymes. Interestingly, the remaining activity of immobilized WaPLA2 onto different supports was more than 50 or 60% after eight recycles or 120 days of storage at 4 °C, respectively. CAC-WaPLA2-II was the best immobilized enzyme complex for the oil degumming process by reducing its final residual phosphorus content from 168 mg/kg to less than 10 mg/kg in only 4 h. Overall, CAC-WaPLA2-II showed the most attractive profiles of temperature, pH, and reaction duration as well as significant storage stability and reusability.
Collapse
Affiliation(s)
- Abir Ben Bacha
- Biochemistry Department,
Science College, King Saud University, P.O. Box 22452, Riyadh 11495, Saudi Arabia
| | - Mona Alonazi
- Biochemistry Department,
Science College, King Saud University, P.O. Box 22452, Riyadh 11495, Saudi Arabia
| |
Collapse
|
5
|
de Oliveira ALN, Lacerda MT, Ramos MJ, Fernandes PA. Viper Venom Phospholipase A2 Database: The Structural and Functional Anatomy of a Primary Toxin in Envenomation. Toxins (Basel) 2024; 16:71. [PMID: 38393149 PMCID: PMC10893444 DOI: 10.3390/toxins16020071] [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: 09/25/2023] [Revised: 01/04/2024] [Accepted: 01/13/2024] [Indexed: 02/25/2024] Open
Abstract
Viper venom phospholipase A2 enzymes (vvPLA2s) and phospholipase A2-like (PLA2-like) proteins are two of the principal toxins in viper venom that are responsible for the severe myotoxic and neurotoxic effects caused by snakebite envenoming, among other pathologies. As snakebite envenoming is the deadliest neglected tropical disease, a complete understanding of these proteins' properties and their mechanisms of action is urgently needed. Therefore, we created a database comprising information on the holo-form, cofactor-bound 3D structure of 217 vvPLA2 and PLA2-like proteins in their physiologic environment, as well as 79 membrane-bound viper species from 24 genera, which we have made available to the scientific community to accelerate the development of new anti-snakebite drugs. In addition, the analysis of the sequenced, 3D structure of the database proteins reveals essential aspects of the anatomy of the proteins, their toxicity mechanisms, and the conserved binding site areas that may anchor universal interspecific inhibitors. Moreover, it pinpoints hypotheses for the molecular origin of the myotoxicity of the PLA2-like proteins. Altogether, this study provides an understanding of the diversity of these toxins and how they are conserved, and it indicates how to develop broad, interspecies, efficient small-molecule inhibitors to target the toxin's many mechanisms of action.
Collapse
Affiliation(s)
| | | | | | - Pedro A. Fernandes
- Requimte-Faculty of Sciences, University of Porto, Rua do Campo Alegre s/n, 4169-000 Porto, Portugal; (A.L.N.d.O.); (M.T.L.); (M.J.R.)
| |
Collapse
|
6
|
Sagar R, Lou J, Best MD. Development of a bis-pyrene phospholipid probe for fluorometric detection of phospholipase A 2 inhibition. Bioorg Med Chem 2023; 87:117301. [PMID: 37150117 PMCID: PMC11070226 DOI: 10.1016/j.bmc.2023.117301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/09/2023]
Abstract
In this work, we report the design, synthesis, and application of a bis-pyrene phospholipid probe for detection of phospholipase A2 action through changes in pyrene monomer and excimer fluorescence intensities. Continuous fluorometric assays enabled detection of the activities of multiple PLA2 enzymes as well as the decrease in catalysis by PLA2 from honey bee venom caused by the inhibitor p-bromo phenacylbromide. Thin-layer chromatography and mass spectrometry analysis were also used to validate probe hydrolysis by PLA2. Mass spectrometry data also supported cleavage of the probe by phospholipase C and D enzymes, although changes in fluorescence were not observed in these cases. Nevertheless, the bis-pyrene phospholipid probe developed in this work is effective for detection of PLA2 enzyme activity through an assay that enables screening for inhibitor development.
Collapse
Affiliation(s)
- Ruhani Sagar
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, TN 37996 USA
| | - Jinchao Lou
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, TN 37996 USA
| | - Michael D Best
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, TN 37996 USA.
| |
Collapse
|
7
|
Castro-Amorim J, Novo de Oliveira A, Da Silva SL, Soares AM, Mukherjee AK, Ramos MJ, Fernandes PA. Catalytically Active Snake Venom PLA 2 Enzymes: An Overview of Its Elusive Mechanisms of Reaction. J Med Chem 2023; 66:5364-5376. [PMID: 37018514 PMCID: PMC10150362 DOI: 10.1021/acs.jmedchem.3c00097] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
Abstract
Snake venom-secreted phospholipase A2 (svPLA2) enzymes, both catalytically active and inactive, are a central component in envenoming. These are responsible for disrupting the cell membrane's integrity, inducing a wide range of pharmacological effects, such as the necrosis of the bitten limb, cardiorespiratory arrest, edema, and anticoagulation. Although extensively characterized, the reaction mechanisms of enzymatic svPLA2 are still to be thoroughly understood. This review presents and analyses the most plausible reaction mechanisms for svPLA2, such as the "single-water mechanism" or the "assisted-water mechanism" initially proposed for the homologous human PLA2. All of the mechanistic possibilities are characterized by a highly conserved Asp/His/water triad and a Ca2+ cofactor. The extraordinary increase in activity induced by binding to a lipid-water interface, known as "interfacial activation," critical for the PLA2s activity, is also discussed. Finally, a potential catalytic mechanism for the postulated noncatalytic PLA2-like proteins is anticipated.
Collapse
Affiliation(s)
- Juliana Castro-Amorim
- LAQV, REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal
| | - Ana Novo de Oliveira
- LAQV, REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal
| | - Saulo Luís Da Silva
- LAQV, REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal
| | - Andreimar M Soares
- Laboratory of Biotechnology of Proteins and Bioactive Compounds (LABIOPROT), Oswaldo Cruz Foundation, National Institute of Epidemiology in the Western Amazon (INCT-EpiAmO), Porto Velho, Rondônia 76812-245, Brazil
- Sao Lucas Universitary Center (UniSL), Porto Velho, Rondônia 76805-846, Brazil
| | - Ashis K Mukherjee
- Microbial Biotechnology and Protein Research Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784028, Assam, India
- Division of Life Sciences, Institute of Advanced Studies in Science and Technology, Vigyan Path, Garchuk, Paschim Boragaon, Guwahati 781035, Assam, India
| | - Maria João Ramos
- LAQV, REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal
| | - Pedro A Fernandes
- LAQV, REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal
| |
Collapse
|
8
|
Molecular Characterization and In Silico Analyses of Maurolipin Structure as a Secretory Phospholipase ( ) from Venom Glands of Iranian Scorpio maurus (Arachnida: Scorpionida). J Trop Med 2022; 2022:1839946. [PMID: 36226273 PMCID: PMC9550507 DOI: 10.1155/2022/1839946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/07/2022] [Accepted: 09/15/2022] [Indexed: 11/18/2022] Open
Abstract
The venom is a mixture of various compounds with specific biological activities, such as the phospholipase A2 (PLA2) enzyme present in scorpion venom. PLA2 plays a key role in inhibiting ryanodine receptor channels and has neurotoxic activity. This study is the first investigation of molecular characterization, cloning, and in silico analyses of PLA2 from Iranian Scorpio maurus, named Maurolipin. After RNA extraction from S. maurus venom glands, cDNA was synthesized and amplified through RT-PCR using specific primers. Amplified Maurolipin was cloned in TA cloning vector, pTG19. For in silico analyses, the characterized gene was analyzed utilizing different software. Maurolipin coding gene with 432 base pair nucleotide length encoded a protein of 144 amino acid residues and 16.34 kilodaltons. Comparing the coding sequence of Maurolipin with other characterized PLA2 from different species of scorpions showed that this protein was a member of the PLA2 superfamily. According to SWISS-MODEL prediction, Maurolipin had 38.83% identity with bee venom PLA2 with 100% confidence and 39% identity with insect phospholipase A2 family, which Phyre2 predicted. According to the three-dimensional structure prediction, Maurolipin with five disulfide bonds has a very high similarity to the structure of PLA2 that belonged to the group III subfamily. The in silico analyses showed that phospholipase A2 coding gene and protein structure is different based on scorpion species and geographical condition in which they live.
Collapse
|
9
|
sPLA2 Wobbles on the Lipid Bilayer between Three Positions, Each Involved in the Hydrolysis Process. Toxins (Basel) 2022; 14:toxins14100669. [PMID: 36287938 PMCID: PMC9610741 DOI: 10.3390/toxins14100669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/14/2022] [Accepted: 09/22/2022] [Indexed: 11/17/2022] Open
Abstract
Secreted phospholipases A2 (sPLA2s) are peripheral membrane enzymes that hydrolyze phospholipids in the sn-2 position. The action of sPLA2 is associated with the work of two active sites. One, the interface binding site (IBS), is needed to bind the enzyme to the membrane surface. The other one, the catalytic site, is needed to hydrolyze the substrate. The interplay between sites, how the substrate protrudes to, and how the hydrolysis products release from, the catalytic site remains in the focus of investigations. Here, we report that bee venom PLA2 has two additional interface binding modes and enzyme activity through constant switching between three different orientations (modes of binding), only one of which is responsible for substrate uptake from the bilayer. The finding was obtained independently using atomic force microscopy and molecular dynamics. Switching between modes has biological significance: modes are steps of the enzyme moving along the membrane, product release in biological milieu, and enzyme desorption from the bilayer surface.
Collapse
|
10
|
Lakshmanan RV, Hull JA, Berry L, Burg M, Bothner B, McKenna R, Agbandje-McKenna M. Structural Dynamics and Activity of B19V VP1u during the pHs of Cell Entry and Endosomal Trafficking. Viruses 2022; 14:1922. [PMID: 36146728 PMCID: PMC9505059 DOI: 10.3390/v14091922] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/28/2022] [Accepted: 08/28/2022] [Indexed: 12/28/2022] Open
Abstract
Parvovirus B19 (B19V) is a human pathogen that is the causative agent of fifth disease in children. It is also known to cause hydrops in fetuses, anemia in AIDS patients, and transient aplastic crisis in patients with sickle cell disease. The unique N-terminus of Viral Protein 1 (VP1u) of parvoviruses, including B19V, exhibits phospholipase A2 (PLA2) activity, which is required for endosomal escape. Presented is the structural dynamics of B19V VP1u under conditions that mimic the pHs of cell entry and endosomal trafficking to the nucleus. Using circular dichroism spectroscopy, the receptor-binding domain of B19V VP1u is shown to exhibit an α-helical fold, whereas the PLA2 domain exhibits a probable molten globule state, both of which are pH invariant. Differential scanning calorimetry performed at endosomal pHs shows that the melting temperature (Tm) of VP1u PLA2 domain is tuned to body temperature (37 °C) at pH 7.4. In addition, PLA2 assays performed at temperatures ranging from 25-45 °C show both a temperature and pH-dependent change in activity. We hypothesize that VP1u PLA2 domain differences in Tm at differing pHs have enabled the virus to "switch on/off" the phospholipase activity during capsid trafficking. Furthermore, we propose the environment of the early endosome as the optimal condition for endosomal escape leading to B19V infection.
Collapse
Affiliation(s)
- Renuk V. Lakshmanan
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, The McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Joshua A. Hull
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, The McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Luke Berry
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59717, USA
| | - Matthew Burg
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, The McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Brian Bothner
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59717, USA
| | - Robert McKenna
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, The McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Mavis Agbandje-McKenna
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, The McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
11
|
Markovic M, Abramov-Harpaz K, Regev C, Ben-Shabat S, Aponick A, Zimmermann EM, Miller Y, Dahan A. Prodrug-Based Targeting Approach for Inflammatory Bowel Diseases Therapy: Mechanistic Study of Phospholipid-Linker-Cyclosporine PLA 2-Mediated Activation. Int J Mol Sci 2022; 23:ijms23052673. [PMID: 35269813 PMCID: PMC8910962 DOI: 10.3390/ijms23052673] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/14/2022] [Accepted: 02/24/2022] [Indexed: 02/04/2023] Open
Abstract
Therapeutics with activity specifically at the inflamed sites throughout the gastrointestinal tract (GIT) would be a major advance in our therapeutic approach to inflammatory bowel disease (IBD). We aimed to develop the prodrug approach that can allow such site-specific drug delivery. Currently, using cyclosporine as a drug of choice in IBD is limited to the most severe cases due to substantial systemic toxicities and narrow therapeutic index of this drug. Previously, we synthesized a series of a phospholipid-linker-cyclosporine (PLC) prodrugs designed to exploit the overexpression of phospholipase A2 (PLA2) in the inflamed intestinal tissues, as the prodrug-activating enzyme. Nevertheless, the extent and rate of prodrug activation differed significantly. In this study we applied in-vitro and modern in-silico tools based on molecular dynamics (MD) simulation, to gain insight into the dynamics and mechanisms of the PLC prodrug activation. We aimed to elucidate the reason for the significant activation change between different linker lengths in our prodrug design. Our work reveals that the PLC conjugate with the 12-carbon linker length yields the optimal prodrug activation by PLA2 in comparison to shorter linker length (6-carbons). This optimized length efficiently allows cyclosporine to be released from the prodrug to the active pocket of PLA2. This newly developed mechanistic approach, presented in this study, can be applied for future prodrug optimization to accomplish optimal prodrug activation and drug targeting in various conditions that include overexpression of PLA2.
Collapse
Affiliation(s)
- Milica Markovic
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (M.M.); (S.B.-S.)
| | - Karina Abramov-Harpaz
- Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (K.A.-H.); (C.R.)
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Clil Regev
- Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (K.A.-H.); (C.R.)
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Shimon Ben-Shabat
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (M.M.); (S.B.-S.)
| | - Aaron Aponick
- Department of Chemistry, University of Florida, Gainesville, FL 32603, USA;
| | - Ellen M. Zimmermann
- Department of Medicine, Division of Gastroenterology, University of Florida, Gainesville, FL 32610, USA;
| | - Yifat Miller
- Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (K.A.-H.); (C.R.)
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Correspondence: (Y.M.); (A.D.)
| | - Arik Dahan
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (M.M.); (S.B.-S.)
- Correspondence: (Y.M.); (A.D.)
| |
Collapse
|
12
|
BthTX-II from Bothrops jararacussu venom has variants with different oligomeric assemblies: An example of snake venom phospholipases A 2 versatility. Int J Biol Macromol 2021; 191:255-266. [PMID: 34547312 DOI: 10.1016/j.ijbiomac.2021.09.083] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 10/20/2022]
Abstract
Phospholipases A2 (PLA2s) are found in almost every venomous snake family. In snakebites, some PLA2s can quickly cause local myonecrosis, which may lead to permanent sequelae if antivenom is administered belatedly. They hydrolyse phospholipids in membranes through a catalytic calcium ions-dependent mechanism. BthTX-II is a basic PLA2 and the second major component in the venom of Bothrops jararacussu. Herein, using the software SEQUENCE SLIDER, which integrates crystallographic, mass spectrometry and genetic data, we characterized the primary, tertiary and quaternary structure of two BthTX-II variants (called a and b), which diverge in 7 residues. Crystallographic structure BthTX-IIa is in a Tense-state with its distorted calcium binding loop buried in the dimer interface, contrarily, the novel BthTX-IIb structure is a monomer in a Relax-state with a fatty acid in the hydrophobic channel. Structural data in solution reveals that both variants are monomeric in neutral physiological conditions and mostly dimeric in an acidic environment, being catalytic active in both situations. Therefore, we propose two myotoxic mechanisms for BthTX-II, a catalytic one associated with the monomeric assembly, whereas the other has a calcium independent activity related to its C-terminal region, adopting a dimeric conformation similar to PLA2-like proteins.
Collapse
|
13
|
Characterization of New Allergens from the Venom of the European Paper Wasp Polistes dominula. Toxins (Basel) 2021; 13:toxins13080559. [PMID: 34437431 PMCID: PMC8402607 DOI: 10.3390/toxins13080559] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/27/2021] [Accepted: 08/06/2021] [Indexed: 01/08/2023] Open
Abstract
Discriminating Polistes dominula and Vespula spp. venom allergy is of growing importance worldwide, as systemic reactions to either species’ sting can lead to severe outcomes. Administering the correct allergen-specific immunotherapy is therefore a prerequisite to ensure the safety and health of venom-allergic patients. Component-resolved diagnostics of Hymenoptera venom allergy might be improved by adding additional allergens to the diagnostic allergen panel. Therefore, three potential new allergens from P. dominula venom—immune responsive protein 30 (IRP30), vascular endothelial growth factor C (VEGF C) and phospholipase A2 (PLA2)—were cloned, recombinantly produced and biochemically characterized. Sera sIgE titers of Hymenoptera venom-allergic patients were measured in vitro to assess the allergenicity and potential cross-reactivity of the venom proteins. IRP30 and VEGF C were classified as minor allergens, as sensitization rates lay around 20–40%. About 50% of P. dominula venom-allergic patients had measurable sIgE titers directed against PLA2 from P. dominula venom. Interestingly, PLA2 was unable to activate basophils of allergic patients, questioning its role in the context of clinically relevant sensitization. Although the obtained results hint to a questionable benefit of the characterized P. dominula venom proteins for improved diagnosis of venom-allergic patients, they can contribute to a deeper understanding of the molecular mechanisms of Hymenoptera venoms and to the identification of factors that determine the allergenic potential of proteins.
Collapse
|
14
|
Barroso IG, Cardoso C, Ferreira C, Terra WR. Transcriptomic and proteomic analysis of the underlying mechanisms of digestion of triacylglycerols and phosphatides and absorption and fate of fatty acids along the midgut of Musca domestica. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2021; 39:100826. [PMID: 33839527 DOI: 10.1016/j.cbd.2021.100826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 03/23/2021] [Accepted: 03/23/2021] [Indexed: 11/24/2022]
Abstract
Most dietary lipids are triacylglycerols (TAGs) and phosphatides that are digested by TAG lipases and phospholipases (PLIPs), respectively, originating fatty acids (FA). The genome of Musca domestica has genes coding for phospholipases A1 (1PLIP), A2 (2PLIP), B (BPLIP), and acid lipases (ALIP), as for proteins involved in activation, binding, and metabolism of FA, which expression in the larval midgut was evaluated by RNA-seq. Some of the codified proteins were identified in midgut microvillar-enriched membrane by proteomics. 1PLIPs are the most expressed PLIPs, mainly in anterior midgut whereas 2PLIPs, and BPLIP in middle and posterior midgut, and ALIPs between middle and posterior regions. Absorption of FAs is putatively accomplished by proteins involved in FA activation (acyl-CoA synthetases) found in microvillar-enriched membrane preparations. Furthermore, FA uptake could be enhanced by proteins that bind FAs (FA-binding proteins) and its activated form (acyl-CoA binding proteins) mainly expressed in posterior midgut. Activated FAs could have different fates: synthesis of diacylglycerol (DAG) and TAG through monoacylglycerol and glycerol-3-phosphate pathways; synthesis of phosphatides; energy source by β-oxidation. Most genes coding for enzymes of those routes is expressed mainly at the end of posterior midgut. Data suggest that phosphatides are digested in anterior midgut by Md1PLIPs, releasing lysophosphatides that emulsify fats to be digested by MdALIPs in the middle and posterior midgut. Most resulting FAs is absorbed in the posterior midgut, where they follow the synthesis of DAG, TAG, and phosphatides or are oxidized along the midgut, mainly in highly metabolic middle and posterior midgut regions.
Collapse
Affiliation(s)
- Ignacio G Barroso
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, 05508-000 São Paulo, Brazil
| | - Christiane Cardoso
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, 05508-000 São Paulo, Brazil
| | - Clelia Ferreira
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, 05508-000 São Paulo, Brazil
| | - Walter R Terra
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, 05508-000 São Paulo, Brazil.
| |
Collapse
|
15
|
Nidamarthi HVK, Choudhury M, Velmurugan D. Understanding the binding mechanism of succinic acid against phospholipase A 2 from bee venom. J Biochem Mol Toxicol 2021; 35:e22715. [PMID: 33580989 DOI: 10.1002/jbt.22715] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 10/19/2020] [Accepted: 11/26/2020] [Indexed: 11/12/2022]
Abstract
Phospholipase A2 (PLA2 ) is responsible for the release of fatty acids from glycerophospholipids. PLA2 is commonly found in mammalian tissues. It is also found in venom from different animals ranging from insects, arachnid, and snakes. The release of arachidonic acid in large amount results in inflammation and pain. Identification of compounds that can inhibit the activity of PLA2 is of large scientific and medicinal interest as these compounds can act as antidotes toward snake bites and bee stings. Among the different compounds that have been tested for inhibition of PLA2 , a secondary metabolite succinic acid is identified to inhibit PLA2 activity. The inhibition was analyzed using an in vitro PLA2 inhibition assay and isothermal titration calorimetry (ITC) studies. The molecular mechanism of the mode of inhibition was studied using molecular docking and simulation studies.
Collapse
Affiliation(s)
- H V Kutumbarao Nidamarthi
- Centre of Advanced Study in Crystallography and Biophysics, University of Madras, Chennai, Tamil Nadu, India
| | - Manisha Choudhury
- Centre of Advanced Study in Crystallography and Biophysics, University of Madras, Chennai, Tamil Nadu, India.,Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, Maharashtra, India
| | - Devadasan Velmurugan
- Centre of Advanced Study in Crystallography and Biophysics, University of Madras, Chennai, Tamil Nadu, India
| |
Collapse
|
16
|
Code C, Ebbesen MF, Sood R, Kinnunen PKJ. Activation of phospholipase A2 by prostaglandin in vitro. Prostaglandins Other Lipid Mediat 2021; 152:106500. [PMID: 33038487 DOI: 10.1016/j.prostaglandins.2020.106500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 09/28/2020] [Accepted: 10/02/2020] [Indexed: 10/23/2022]
Abstract
Prostaglandins are a diverse family of biological active molecules that are synthesized after liberation of arachnidonic or linolenic acid from the plasma membrane by phospholipase A2 (PLA2). Specific prostaglandins may be pro-inflammatory or anti-inflammatory due to a poorly understood biochemical equilibrium. Some of the anti-inflammatory prostaglandins namely, prostaglandin A1 (PGA1) and prostaglandin E1 (PGE1) have a cyclopentenone moiety that can react and modify a protein's activity. These two prostaglandins are electrophilic reactive lipid species and are formed as a result of the reaction cascade initiated by PLA2. It was of interest to study the interaction with these prostaglandins as they could either amplify or block this enzyme's activity. We found that the former is true initially as there is a shorter time to activate the protein on the lipid membrane and an overall increase in hydrolysis was observed when PGA1 and PGE1 prostaglandin was added with PLA2 and liposomes. The interfacial activation model was further explored in which there is a modification of the enzyme rather than a modifcation of the substrate. However, after a time the protein was shown to form amyloid like fibrils thereby blocking further hydrolysis. The fibrillization kinetics in the presence of the one of the prostaglandins was monitored using thioflavin T (ThT) and the resulting fibrils were characterized using transmission electron microscopy (TEM) and atomic force microscopy (AFM). Modification of PLA2 by these prostaglandins leading to amyloid like fibrils gives an additional perspective of control of the interfacial activation mechanism of this enzyme.
Collapse
Affiliation(s)
- Christian Code
- Institute of Biomedical Engineering and Computational Science, Aalto University, Espoo, Finland; Centre for Single Particle Science and Engineering, MEMPHYS, University of Southern Denmark, Odense, Denmark
| | - Morten Frendø Ebbesen
- Centre for Single Particle Science and Engineering, MEMPHYS, University of Southern Denmark, Odense, Denmark
| | - Rohit Sood
- Institute of Biomedical Engineering and Computational Science, Aalto University, Espoo, Finland; Department of Medical Biochemistry and Genetics, Faculty of Medicine, University of Turku, Turku, Finland; Spinverse Oy, Tekniikantie 14, 02150, Espoo, Finland.
| | - Paavo K J Kinnunen
- Institute of Biomedical Engineering and Computational Science, Aalto University, Espoo, Finland
| |
Collapse
|
17
|
Darwish DA, Masoud HMM, Abdel-Monsef MM, Helmy MS, Zidan HA, Ibrahim MA. Phospholipase A2 enzyme from the venom of Egyptian honey bee Apis mellifera lamarckii with anti-platelet aggregation and anti-coagulation activities. J Genet Eng Biotechnol 2021; 19:10. [PMID: 33443641 PMCID: PMC7809086 DOI: 10.1186/s43141-020-00112-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/28/2020] [Indexed: 11/24/2022]
Abstract
Background Honey bee venom contains various enzymes with wide medical and pharmaceutical applications. Results The phospholipase A2 (PLA2) has been apparently purified from the venom of Egyptian honey bee (Apis mellifera lamarckii) 8.9-fold to a very high specific activity of 6033 U/mg protein using DEAE–cellulose and Sephacryl S-300 columns. The purified bee venom PLA2 is monomeric 16 kDa protein and has isoelectric point (pI) of 5.9. The optimal activity of bee venom PLA2 was attained at pH 8 and 45 °C. Cu2+, Ni2+, Fe2+, Ca2+, and Co2+ exhibited a complete activating effect on it, while Zn2+, Mn2+, NaN3, PMSF, N-Methylmaleimide, and EDTA have inhibitory effect. Conclusions The purified bee venom PLA2 exhibited anti-platelet aggregation and anti-coagulation activities which makes it promising agent for developing novel anti-clot formation drugs in future.
Collapse
Affiliation(s)
- Doaa A Darwish
- Molecular Biology Department, National Research Centre, 33 El Bohouth St. (former El Tahrir St.), Dokki, P.O. 12622, Giza, Egypt
| | - Hassan M M Masoud
- Molecular Biology Department, National Research Centre, 33 El Bohouth St. (former El Tahrir St.), Dokki, P.O. 12622, Giza, Egypt.
| | - Mohamed M Abdel-Monsef
- Molecular Biology Department, National Research Centre, 33 El Bohouth St. (former El Tahrir St.), Dokki, P.O. 12622, Giza, Egypt
| | - Mohamed S Helmy
- Molecular Biology Department, National Research Centre, 33 El Bohouth St. (former El Tahrir St.), Dokki, P.O. 12622, Giza, Egypt
| | - Hind A Zidan
- Plant Protection Research Institute, Agricultural Research Center, Giza, Egypt
| | - Mahmoud A Ibrahim
- Molecular Biology Department, National Research Centre, 33 El Bohouth St. (former El Tahrir St.), Dokki, P.O. 12622, Giza, Egypt
| |
Collapse
|
18
|
A New Group II Phospholipase A2 from Walterinnesia aegyptia Venom with Antimicrobial, Antifungal, and Cytotoxic Potential. Processes (Basel) 2020. [DOI: 10.3390/pr8121560] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Many venomous species, especially snakes, contain a variety of secreted phospholipases A2 that contribute to venom toxicity and prey digestion. We characterized a novel highly toxic phospholipase A2 of group II, WaPLA2-II, from the snake venom of Saudi Walterinnesia aegyptia (W. aegyptia). The enzyme was purified using a reverse phase C18 column. It is a monomeric protein with a molecular weight of approximately 14 kDa and an NH2-terminal amino acid sequence exhibiting similarity to the PLA2 group II enzymes. WaPLA2-II, which contains 2.5% (w/w) glycosylation, reached a maximal specific activity of 1250 U/mg at pH 9.5 and 55 °C in the presence of Ca2+ and bile salts. WaPLA2-II was also highly stable over a large pH and temperature range. A strong correlation between antimicrobial and indirect hemolytic activities of WaPLA2 was observed. Additionally, WaPLA2-II was found to be significantly cytotoxic only on cancerous cells. However, chemical modification with para-Bromophenacyl bromide (p-BPB) inhibited WaPLA2-II enzymatic activity without affecting its antitumor effect, suggesting the presence of a separate ‘pharmacological site’ in snake venom phospholipase A2 via its receptor binding affinity. This enzyme is a candidate for applications including the treatment of phospholipid-rich industrial effluents and for the food production industry. Furthermore, it may represent a new therapeutic lead molecule for treating cancer and microbial infections.
Collapse
|
19
|
Alekseeva AS, Volynsky PE, Krylov NA, Chernikov VP, Vodovozova EL, Boldyrev IA. Phospholipase A2 way to hydrolysis: Dint formation, hydrophobic mismatch, and lipid exclusion. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1863:183481. [PMID: 33002451 DOI: 10.1016/j.bbamem.2020.183481] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/23/2020] [Accepted: 09/21/2020] [Indexed: 01/05/2023]
Abstract
Phospholipase A2 (PLA2) exerts a wide range of biological effects and attracts a lot of attention of researchers. Two sites are involved in manifestation of PLA2 enzymatic activity: catalytic site responsible for substrate binding and fatty acid cleavage from the sn-2 position of a glycerophospholipid, and interface binding site (IBS) responsible for the protein binding to lipid membrane. IBS is formed by positively charged and hydrophobic amino acids on the outer surface of the protein molecule. Understanding the mechanism of PLA2 interaction with the lipid membrane is the most challenging step in biochemistry of this enzyme. We used a combination of experimental and computer simulation techniques to clarify molecular details of bee venom PLA2 interaction with lipid bilayers formed by palmitoyloleoylphosphatidylcholine or dipalmitoylphosphatidylcholine. We found that after initial enzyme contact with the membrane, a network of hydrogen bonds was formed. This led to deformation of the interacting leaflet and dint formation. The bilayer response to the deformation depended on its phase state. In a gel-phase bilayer, diffusion of lipids is restricted therefore chain melting occurred in both leaflets of the bilayer. In the case of a fluid-phase bilayer, lateral diffusion is possible, and lipid polar head groups were excluded from the contact area. As a result, the bilayer became thinner and a large hydrophobic area was formed. We assume that relative ability of a bilayer to come through lipid redistribution process defines the rate of initial stages of the catalysis.
Collapse
Affiliation(s)
- Anna S Alekseeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya st., 16/10, 117997 Moscow, Russia
| | - Pavel E Volynsky
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya st., 16/10, 117997 Moscow, Russia
| | - Nikolay A Krylov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya st., 16/10, 117997 Moscow, Russia
| | - Valery P Chernikov
- Scientific Research Institute of Human Morphology, Tsyurupy st., 3, 117418 Moscow, Russia
| | - Elena L Vodovozova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya st., 16/10, 117997 Moscow, Russia
| | - Ivan A Boldyrev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya st., 16/10, 117997 Moscow, Russia.
| |
Collapse
|
20
|
Zahirović A, Luzar J, Molek P, Kruljec N, Lunder M. Bee Venom Immunotherapy: Current Status and Future Directions. Clin Rev Allergy Immunol 2020; 58:326-341. [PMID: 31240545 DOI: 10.1007/s12016-019-08752-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Bee venom immunotherapy is the main treatment option for bee sting allergy. Its major limitations are the high percentage of allergic side effects and long duration, which are driving the development of novel therapeutic modalities. Three general approaches have been evaluated including the use of hypoallergenic allergen derivatives, adjunctive therapy, and alternative delivery routes. This article reviews preclinical and clinical evidence on the therapeutic potential of these new therapies. Among hypoallergenic derivatives, hybrid allergens showed a markedly reduced IgE reactivity in mouse models. Whether they will offer therapeutic benefit over extract, it is still not known since clinical trials have not been carried out yet. T cell epitope peptides have proven effective in small clinical trials. Major histocompatibility complex class II restriction was circumvented by using long overlapping or promiscuous T cell epitope peptides. However, the T cell-mediated late-phase adverse events have been reported with both short and longer peptides. Application of mimotopes could potentially overcome both T cell- and IgE-mediated adverse events. During this evolution of vaccine, there has been a gain in safety. The efficacy was further improved with the use of Toll-like receptor-activating adjuvants and delivery systems. In murine models, the association of allergen Api m 1 with cytosine-guanosine rich oligonucleotides stimulated strong T-helper type-1 response, whereas its encapsulation into microbubbles protected mice against allergen challenge. An intralymphatic administration of low-dose vaccine has shown the potential to decrease treatment from 5 years to only 12 weeks. Bigger clinical trials are needed to follow up on these results.
Collapse
Affiliation(s)
- Abida Zahirović
- Faculty of Pharmacy, Department of Pharmaceutical Biology, University of Ljubljana, Aškerčeva 7, SI-1000, Ljubljana, Slovenia.
| | - Jernej Luzar
- Faculty of Pharmacy, Department of Pharmaceutical Biology, University of Ljubljana, Aškerčeva 7, SI-1000, Ljubljana, Slovenia
| | - Peter Molek
- Faculty of Pharmacy, Department of Pharmaceutical Biology, University of Ljubljana, Aškerčeva 7, SI-1000, Ljubljana, Slovenia
| | - Nika Kruljec
- Faculty of Pharmacy, Department of Pharmaceutical Biology, University of Ljubljana, Aškerčeva 7, SI-1000, Ljubljana, Slovenia
| | - Mojca Lunder
- Faculty of Pharmacy, Department of Pharmaceutical Biology, University of Ljubljana, Aškerčeva 7, SI-1000, Ljubljana, Slovenia
| |
Collapse
|
21
|
An Integrated Proteomic and Transcriptomic Analysis Reveals the Venom Complexity of the Bullet Ant Paraponera clavata. Toxins (Basel) 2020; 12:toxins12050324. [PMID: 32422990 PMCID: PMC7290781 DOI: 10.3390/toxins12050324] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 01/11/2023] Open
Abstract
A critical hurdle in ant venom proteomic investigations is the lack of databases to comprehensively and specifically identify the sequence and function of venom proteins and peptides. To resolve this, we used venom gland transcriptomics to generate a sequence database that was used to assign the tandem mass spectrometry (MS) fragmentation spectra of venom peptides and proteins to specific transcripts. This was performed alongside a shotgun liquid chromatography-mass spectrometry (LC-MS/MS) analysis of the venom to confirm that these assigned transcripts were expressed as proteins. Through the combined transcriptomic and proteomic investigation of Paraponera clavata venom, we identified four times the number of proteins previously identified using 2D-PAGE alone. In addition to this, by mining the transcriptomic data, we identified several novel peptide sequences for future pharmacological investigations, some of which conform with inhibitor cysteine knot motifs. These types of peptides have the potential to be developed into pharmaceutical or bioinsecticide peptides.
Collapse
|
22
|
Ultra-high throughput single-cell analysis of proteins and RNAs by split-pool synthesis. Commun Biol 2020; 3:213. [PMID: 32382044 PMCID: PMC7205613 DOI: 10.1038/s42003-020-0896-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 03/04/2020] [Indexed: 12/11/2022] Open
Abstract
Single-cell omics provide insight into cellular heterogeneity and function. Recent technological advances have accelerated single-cell analyses, but workflows remain expensive and complex. We present a method enabling simultaneous, ultra-high throughput single-cell barcoding of millions of cells for targeted analysis of proteins and RNAs. Quantum barcoding (QBC) avoids isolation of single cells by building cell-specific oligo barcodes dynamically within each cell. With minimal instrumentation (four 96-well plates and a multichannel pipette), cell-specific codes are added to each tagged molecule within cells through sequential rounds of classical split-pool synthesis. Here we show the utility of this technology in mouse and human model systems for as many as 50 antibodies to targeted proteins and, separately, >70 targeted RNA regions. We demonstrate that this method can be applied to multi-modal protein and RNA analyses. It can be scaled by expansion of the split-pool process and effectively renders sequencing instruments as versatile multi-parameter flow cytometers. Maeve O’Huallachain et al. report a method that enables simultaneous, ultra-high throughput single-cell barcoding for targeted single-cell protein and RNA analysis. They show the utility of their method in analyses of mRNA and protein expression in human and mouse cells.
Collapse
|
23
|
Smichi N, Othman H, Zarai Z, Fendri A, Abousalham A. -Identification of a novel intestinal phospholipase A2 from annular seabream: Insights into its catalytic mechanism and its role in biological processes. Process Biochem 2020. [DOI: 10.1016/j.procbio.2019.12.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
24
|
Zhang F, Li X, Ma Y, Wang C, Hu P, Wang F, Lu X. Illustrating Interfacial Interaction between Honey Bee Venom Phospholipase A 2 and Supported Negatively Charged Lipids with Sum Frequency Generation and Laser Scanning Confocal Microscopy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:2946-2953. [PMID: 32093479 DOI: 10.1021/acs.langmuir.0c00003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Phospholipase A2 is an important enzyme species which can widely be found in animals, plants, bacteria, and so on. A large number of studies have shown that phospholipase A2 is highly catalytic toward the lipids. Here, sum frequency generation (SFG) vibrational spectroscopy and laser scanning confocal microscopy (LSCM) were applied to study the interaction between honey bee venom phospholipase A2 (bvPLA2) and the negatively charged DPPG bilayer. In both cases without and with the calcium ions (Ca2+), the bvPLA2 molecules were adsorbed onto the outer leaflet surface with the orientational order, and the adsorbed bvPLA2 molecules damaged the order of the packed outer leaflet. In comparison to the case without Ca2+, the addition of Ca2+ can accelerate the attaching process of bvPLA2 to the outer leaflet surface and decelerate the process of damaging the outer leaflet order. The experimental result also confirmed, with the help of the Ca2+, the DPPG molecules in the outer leaflet were hydrolyzed, with both hydrolysates, that is, lysophospholipids and fatty acids, remaining at the interface, showing a distinct difference from previous published literatures regarding neutral lipids [Phys. Chem. Chem. Phys. 2018, 20, 63-67] and PLA1 [Langmuir 2019, 35, 12831-12838].
Collapse
Affiliation(s)
- Furong Zhang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Xu Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Yonghao Ma
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Chu Wang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Pengcheng Hu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Feng Wang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Xiaolin Lu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| |
Collapse
|
25
|
Tjørnelund HD, Madsen JJ, Peters GHJ. Water-Intake and Water-Molecule Paths to the Active Site of Secretory Phospholipase A 2 Studied Using MD Simulations and the Tracking Tool AQUA-DUCT. J Phys Chem B 2020; 124:1881-1891. [PMID: 32064878 DOI: 10.1021/acs.jpcb.9b10837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Secretory phospholipases A2 (sPLA2s) are a subclass of enzymes that catalyze the hydrolysis at the sn-2 position of glycerophospholipids, producing free fatty acids and lysophospholipids. In this study, different phospholipids with structural modifications close to the scissile sn-2 ester bond were studied to determine the effect of the structural changes on the formation of the Michaelis-Menten complex and the water entry/exit pathways using molecular dynamics simulations and the computational tracking tool AQUA-DUCT. Structural modifications include methylation, dehydrogenation, and polarization close to the sn-2 scissile bond. We found that all water molecules reaching the active site of sPLA2-IIA pass by the aromatic residues Phe5 and Tyr51 and enter the active site through an active-site cleft. The relative amount of water available for the enzymatic reaction of the different phospholipid-sPLA2 complexes was determined together with the distance between key atoms in the catalytic machinery. The results showed that (Z)-unsaturated phospholipid is a good substrate for sPLA2-IIA. The computational results are in good agreement with previously reported experimental data on the ability of sPLA2-IIA to hydrolyze liposomes made from the different phospholipids, and the results provide new insights into the necessary active-site solvation of the Michaelis-Menten complex and can pave the road for rational design in engineering applications.
Collapse
Affiliation(s)
- Helena D Tjørnelund
- Department of Chemistry, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| | - Jesper J Madsen
- Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida 33620, United States
| | - Günther H J Peters
- Department of Chemistry, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| |
Collapse
|
26
|
Cardoso FF, Gomes AAS, Dreyer TR, Cavalcante WLG, Dal Pai M, Gallacci M, Fontes MRM. Neutralization of a bothropic PLA 2-like protein by caftaric acid, a novel potent inhibitor of ophidian myotoxicity. Biochimie 2020; 170:163-172. [PMID: 31978419 DOI: 10.1016/j.biochi.2020.01.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 01/19/2020] [Indexed: 11/18/2022]
Abstract
Envenoming by snakebite is an important global health issue that has received little attention, leading the World Health Organization to naming it as neglected tropical disease. Several snakebites present serious local symptoms manifested on victims that may not be efficiently neutralized by serum therapy. Phospholipase A2-like (PLA2-like) toxins are present in Viperidae venoms and are responsible for local myotoxic activity. Herein, we investigated the association between BthTX-I toxin and caftaric acid (CFT), a molecule present in plants. CFT neutralized neuromuscular blocking and muscle-damaging activities promoted by BthTX-I. Calorimetric and light-scattering assays demonstrated that CFT inhibitor interacted with dimeric BthTX-I. Bioinformatics simulations indicated that CFT inhibitor binds to the toxin's hydrophobic channel (HCh). According to the current myotoxic mechanism, three different regions of PLA2-like toxins have specific tasks: protein allosteric activation (HCh), membrane dockage (MDoS), and membrane rupture (MDiS). We propose CFT inhibitor interferes with the allosteric activation, which is related to the conformation change leading to the exposure/alignment of MDoS/MDiS region. This is the first report of a PLA2-like toxin fully inhibited by a compound that interacts only with its HCh region. Thus, CFT is a novel candidate to complement serum therapy and improve the treatment of snakebite.
Collapse
Affiliation(s)
- Fábio F Cardoso
- Departamento de Física e Biofísica, Instituto de Biociências, Universidade Estadual Paulista (UNESP), Botucatu, SP, Brazil
| | - Antoniel A S Gomes
- Departamento de Física e Biofísica, Instituto de Biociências, Universidade Estadual Paulista (UNESP), Botucatu, SP, Brazil
| | - Thiago R Dreyer
- Departamento de Física e Biofísica, Instituto de Biociências, Universidade Estadual Paulista (UNESP), Botucatu, SP, Brazil
| | - Walter L G Cavalcante
- Departamento de Física e Biofísica, Instituto de Biociências, Universidade Estadual Paulista (UNESP), Botucatu, SP, Brazil; Departamento de Farmacologia, Instituto de Biociências, Universidade Estadual Paulista (UNESP), Botucatu, SP, Brazil
| | - Maeli Dal Pai
- Departamento de Morfologia, Instituto de Biociências, Universidade Estadual Paulista (UNESP), Botucatu, SP, Brazil
| | - Márcia Gallacci
- Departamento de Farmacologia, Instituto de Biociências, Universidade Estadual Paulista (UNESP), Botucatu, SP, Brazil
| | - Marcos R M Fontes
- Departamento de Física e Biofísica, Instituto de Biociências, Universidade Estadual Paulista (UNESP), Botucatu, SP, Brazil.
| |
Collapse
|
27
|
Zhang P, Villanueva V, Kalkowski J, Liu C, Donovan AJ, Bu W, Schlossman ML, Lin B, Liu Y. Molecular interactions of phospholipid monolayers with a model phospholipase. SOFT MATTER 2019; 15:4068-4077. [PMID: 30958491 DOI: 10.1039/c8sm01154k] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The intrinsic overexpression of secretory phospholipase A2 (sPLA2) in various pro-inflammatory diseases and cancers has the potential to be exploited as a therapeutic strategy for diagnostics and treatment. To explore this potential and advance our knowledge of the role of sPLA2 in related diseases, it is necessary to systematically investigate the molecular interaction of the enzyme with lipids. By employing a Langmuir trough integrated with X-ray reflectivity and grazing incidence X-ray diffraction techniques, this study examined the molecular packing structure of 1,2-palmitoyl-sn-glycero-3-phosphocholine (DPPC) films before and after enzyme adsorption and enzyme-catalyzed degradation. Molecular interaction of sPLA2 (from bee venom) with the DPPC monolayer exhibited Ca2+ dependence. DPPC molecules at the interface without Ca2+ retained a monolayer organization; upon adsorption of sPLA2 to the monolayer the packing became tighter. In contrast, sPLA2-catalyzed degradation of DPPC occurred in the presence of Ca2+, leading to disruption of the ordered monolayer structure of DPPC. The interfacial film became a mixture of highly ordered multilayer domains of palmitic acid (PA) and loosely packed monolayer phase of 1-palmitoyl-2-hydroxy-sn-glycero-3-phosphocholine (lysoPC) that potentially contained the remaining un-degraded DPPC. The redistribution of lipid degradation products into the third dimension, which produced multilayer PA domains, damaged the structural integrity of the original lipid layer and may explain the bursting of liposomes observed in other studies after a latency period of mixing liposomes with sPLA2. A quantitative understanding of the lipid packing and lipid-enzyme interaction provides an intuitive means of designing and optimizing lipid-related drug delivery systems.
Collapse
Affiliation(s)
- Pin Zhang
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Markovic M, Dahan A, Keinan S, Kurnikov I, Aponick A, Zimmermann EM, Ben-Shabat S. Phospholipid-Based Prodrugs for Colon-Targeted Drug Delivery: Experimental Study and In-Silico Simulations. Pharmaceutics 2019; 11:pharmaceutics11040186. [PMID: 30995772 PMCID: PMC6523355 DOI: 10.3390/pharmaceutics11040186] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/07/2019] [Accepted: 04/08/2019] [Indexed: 12/23/2022] Open
Abstract
In ulcerative colitis (UC), the inflammation is localized in the colon, and one of the successful strategies for colon-targeting drug delivery is the prodrug approach. In this work, we present a novel phospholipid (PL)-based prodrug approach, as a tool for colonic drug targeting in UC. We aim to use the phospholipase A2 (PLA2), an enzyme that is overexpressed in the inflamed colonic tissues of UC patients, as the PL-prodrug activating enzyme, to accomplish the liberation of the parent drug from the prodrug complex at the specific diseased tissue(s). Different linker lengths between the PL and the drug moiety can dictate the rate of activation by PLA2, and subsequently determine the amount of free drugs at the site of action. The feasibility of this approach was studied with newly synthesized PL-Fmoc (fluorenylmethyloxycarbonyl) conjugates, using Fmoc as a model compound for testing our hypothesis. In vitro incubation with bee venom PLA2 demonstrated that a 7-carbon linker between the PL and Fmoc has higher activation rate than a 5-carbon linker. 4-fold higher colonic expression of PLA2 was demonstrated in colonic mucosa of colitis-induced rats when compared to healthy animals, validating our hypothesis of a colitis-targeting prodrug approach. Next, a novel molecular dynamics (MD) simulation was developed for PL-based prodrugs containing clinically relevant drugs. PL-methotrexate conjugate with 6-carbon linker showed the highest extent of PLA2-mediated activation, whereas shorter linkers were activated to a lower extent. In conclusion, this work demonstrates that for carefully designed PL-drug conjugates, PLA2 overexpression in inflamed colonic tissues can be used as prodrug-activating enzyme and drug targeting strategy, including insights into the activation mechanisms in a PLA2 binding site.
Collapse
Affiliation(s)
- Milica Markovic
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel.
| | - Arik Dahan
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel.
| | | | | | - Aaron Aponick
- Department of Chemistry, University of Florida, Gainesville, FL 32603, USA.
| | - Ellen M Zimmermann
- Department of Medicine, Division of Gastroenterology, University of Florida, Gainesville, FL 32603, USA.
| | - Shimon Ben-Shabat
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel.
| |
Collapse
|
29
|
The Dual Prey-Inactivation Strategy of Spiders-In-Depth Venomic Analysis of Cupiennius salei. Toxins (Basel) 2019; 11:toxins11030167. [PMID: 30893800 PMCID: PMC6468893 DOI: 10.3390/toxins11030167] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 03/14/2019] [Indexed: 02/08/2023] Open
Abstract
Most knowledge of spider venom concerns neurotoxins acting on ion channels, whereas proteins and their significance for the envenomation process are neglected. The here presented comprehensive analysis of the venom gland transcriptome and proteome of Cupiennius salei focusses on proteins and cysteine-containing peptides and offers new insight into the structure and function of spider venom, here described as the dual prey-inactivation strategy. After venom injection, many enzymes and proteins, dominated by α-amylase, angiotensin-converting enzyme, and cysteine-rich secretory proteins, interact with main metabolic pathways, leading to a major disturbance of the cellular homeostasis. Hyaluronidase and cytolytic peptides destroy tissue and membranes, thus supporting the spread of other venom compounds. We detected 81 transcripts of neurotoxins from 13 peptide families, whereof two families comprise 93.7% of all cysteine-containing peptides. This raises the question of the importance of the other low-expressed peptide families. The identification of a venom gland-specific defensin-like peptide and an aga-toxin-like peptide in the hemocytes offers an important clue on the recruitment and neofunctionalization of body proteins and peptides as the origin of toxins.
Collapse
|
30
|
Perez-Riverol A, Lasa AM, Dos Santos-Pinto JRA, Palma MS. Insect venom phospholipases A1 and A2: Roles in the envenoming process and allergy. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2019; 105:10-24. [PMID: 30582958 DOI: 10.1016/j.ibmb.2018.12.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 12/10/2018] [Accepted: 12/18/2018] [Indexed: 06/09/2023]
Abstract
Insect venom phospholipases have been identified in nearly all clinically relevant social Hymenoptera, including bees, wasps and ants. Among other biological roles, during the envenoming process these enzymes cause the disruption of cellular membranes and induce hypersensitive reactions, including life threatening anaphylaxis. While phospholipase A2 (PLA2) is a predominant component of bee venoms, phospholipase A1 (PLA1) is highly abundant in wasps and ants. The pronounced prevalence of IgE-mediated reactivity to these allergens in sensitized patients emphasizes their important role as major elicitors of Hymenoptera venom allergy (HVA). PLA1 and -A2 represent valuable marker allergens for differentiation of genuine sensitizations to bee and/or wasp venoms from cross-reactivity. Moreover, in massive attacks, insect venom phospholipases often cause several pathologies that can lead to fatalities. This review summarizes the available data related to structure, model of enzymatic activity and pathophysiological roles during envenoming process of insect venom phospholipases A1 and -A2.
Collapse
Affiliation(s)
- Amilcar Perez-Riverol
- Center of the Study of Social Insects, Department of Biology, Institute of Biosciences of Rio Claro, São Paulo State University (UNESP), Rio Claro, SP, 13500, Brazil
| | - Alexis Musacchio Lasa
- Center for Genetic Engineering and Biotechnology, Biomedical Research Division, Department of System Biology, Ave. 31, e/158 and 190, P.O. Box 6162, Cubanacan, Playa, Havana, 10600, Cuba
| | - José Roberto Aparecido Dos Santos-Pinto
- Center of the Study of Social Insects, Department of Biology, Institute of Biosciences of Rio Claro, São Paulo State University (UNESP), Rio Claro, SP, 13500, Brazil
| | - Mario Sergio Palma
- Center of the Study of Social Insects, Department of Biology, Institute of Biosciences of Rio Claro, São Paulo State University (UNESP), Rio Claro, SP, 13500, Brazil.
| |
Collapse
|
31
|
Structural diversity of arthropod venom toxins. Toxicon 2018; 152:46-56. [DOI: 10.1016/j.toxicon.2018.07.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 06/29/2018] [Accepted: 07/19/2018] [Indexed: 11/19/2022]
|
32
|
dos Santos-Pinto JRA, Perez-Riverol A, Lasa AM, Palma MS. Diversity of peptidic and proteinaceous toxins from social Hymenoptera venoms. Toxicon 2018; 148:172-196. [DOI: 10.1016/j.toxicon.2018.04.029] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/24/2018] [Accepted: 04/25/2018] [Indexed: 12/20/2022]
|
33
|
Krayem N, Parsiegla G, Gaussier H, Louati H, Jallouli R, Mansuelle P, Carrière F, Gargouri Y. Functional characterization and FTIR-based 3D modeling of full length and truncated forms of Scorpio maurus venom phospholipase A 2. Biochim Biophys Acta Gen Subj 2018; 1862:1247-1261. [DOI: 10.1016/j.bbagen.2018.02.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 01/26/2018] [Accepted: 02/06/2018] [Indexed: 11/24/2022]
|
34
|
Efficient heterologous expression, functional characterization and molecular modeling of annular seabream digestive phospholipase A2. Chem Phys Lipids 2018. [DOI: 10.1016/j.chemphyslip.2017.06.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
35
|
Secreted Phospholipases A₂ from Animal Venoms in Pain and Analgesia. Toxins (Basel) 2017; 9:toxins9120406. [PMID: 29311537 PMCID: PMC5744126 DOI: 10.3390/toxins9120406] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/13/2017] [Accepted: 12/16/2017] [Indexed: 12/20/2022] Open
Abstract
Animal venoms comprise a complex mixture of components that affect several biological systems. Based on the high selectivity for their molecular targets, these components are also a rich source of potential therapeutic agents. Among the main components of animal venoms are the secreted phospholipases A2 (sPLA2s). These PLA2 belong to distinct PLA2s groups. For example, snake venom sPLA2s from Elapidae and Viperidae families, the most important families when considering envenomation, belong, respectively, to the IA and IIA/IIB groups, whereas bee venom PLA2 belongs to group III of sPLA2s. It is well known that PLA2, due to its hydrolytic activity on phospholipids, takes part in many pathophysiological processes, including inflammation and pain. Therefore, secreted PLA2s obtained from animal venoms have been widely used as tools to (a) modulate inflammation and pain, uncovering molecular targets that are implicated in the control of inflammatory (including painful) and neurodegenerative diseases; (b) shed light on the pathophysiology of inflammation and pain observed in human envenomation by poisonous animals; and, (c) characterize molecular mechanisms involved in inflammatory diseases. The present review summarizes the knowledge on the nociceptive and antinociceptive actions of sPLA2s from animal venoms, particularly snake venoms.
Collapse
|
36
|
Tilinca M, Florea A. Ultrastructural analysis of early toxic effects produced by bee venom phospholipase A2 and melittin in Sertoli cells in rats. Toxicon 2017; 141:94-103. [PMID: 29229236 DOI: 10.1016/j.toxicon.2017.12.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 11/15/2017] [Accepted: 12/06/2017] [Indexed: 10/18/2022]
Abstract
In this study, we aimed to investigate the testicular toxicity of two molecules derived from bee venom (BV): phospholipase A2 (PlA2) and melittin (Mlt). Ultrastructural effects of purified BV PlA2 and Mlt were assessed consecutive to repeated dose (30 days) and acute toxicity studies. For the subchronic treatment, PlA2 and Mlt were injected in daily doses equivalent to those released by a bee sting (105 μg PlA2/kg/day and 350 μg Mlt/kg/day), while in the acute treatment their doses corresponded to those released by 100 bee stings (9.3 mg PlA2/kg and 31 mg Mlt/kg). Both PlA2 and Mlt affected the Leydig cells and the cells in seminiferous tubules, the Sertoli cells first of all. PlA2 injection resulted in detachment of the Sertoli cells from the surrounding cells, and extracellular vacuolations, cytoplasmic vacuolations in their basal region and in branches as well, detachment of spermatids, residual bodies and sometimes even spermatocytes into the lumen, changes that had a higher magnitude after the acute treatment. Mlt injection induced similar ultrastructural alterations, but more severe, including degeneration of cellular organelles and cellular necrosis, resulting into rarefaction of the seminiferous epithelium; the ultrastructural changes had a higher magnitude after the 30 repeated dose treatment. We concluded that either of the two molecules tested here, PlA2 and Mlt, were Sertoli cells toxicants at the used doses, and they participated both in the BV testicular toxicity. We consider the observed changes as part of a preceding mechanism of the more severe alterations produced by the BV. It also remains possible that these early unspecific changes reported here could represent the response of the SCs not only to the components of bee venom, but to molecules of other venoms as well. The Sertoli cells were the primary target of PlA2 and Mlt in the spermatogenic epithelium, and their alteration led to further degenerative changes of the germ cells. Since the exposure to PlA2 and Mlt caused severe alteration, including cell death and detachment of immature germ cells into the lumen, we may also conclude that the bee venom molecules had a potential to interfere with normal progression of spermatogenesis. All the degenerative changes observed in the Sertoli cells were accompanied with changes of the Leydig cells.
Collapse
Affiliation(s)
- Mariana Tilinca
- Department of Cell and Molecular Biology, Faculty of Medicine, University of Medicine and Pharmacy, Târgu-Mureş, Romania
| | - Adrian Florea
- Department of Cell and Molecular Biology, Faculty of Medicine, "Iuliu Haţieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania.
| |
Collapse
|
37
|
Smichi N, Othman H, Achouri N, Noiriel A, Arondel V, Srairi-Abid N, Abousalham A, Gargouri Y, Miled N, Fendri A. Functional and Structural Characterization of a Thermostable Phospholipase A 2 from a Sparidae Fish (Diplodus annularis). JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:2468-2480. [PMID: 28287729 DOI: 10.1021/acs.jafc.6b05810] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Novel phospholipase (PLA2) genes from the Sparidae family were cloned. The sequenced PLA2 revealed an identity with pancreatic PLA2 group IB. To better understand the structure/function relationships of these enzymes and their evolution, the Diplodus annularis PLA2 (DaPLA2) was overexpressed in E. coli. The refolded enzyme was purified by Ni-affinity chromatography and has a molecular mass of 15 kDa as determined by MALDI-TOF spectrometry. Interestingly, unlike the pancreatic type, the DaPLA2 was active and stable at higher temperatures, which suggests its great potential in biotechnological applications. The 3D structure of DaPLA2 was constructed to gain insights into the functional properties of sparidae PLA2. Molecular docking and dynamic simulations were performed to explain the higher thermal stability and the substrate specificity of DaPLA2. Using the monolayer technique, the purified DaPLA2 was found to be active on various phospholipids ranging from 10 to 20 mN·m-1, which explained the absence of the hemolytic activity for DaPLA2.
Collapse
Affiliation(s)
- Nabil Smichi
- Laboratory of Biochemistry and Enzymatic Engineering of Lipases, ENIS , 3038 Sfax, Tunisia
- Enzymologie Interfaciale et Physiologie de la Lipolyse, UMR7282, CNRS, Aix-Marseille Université , 31 Chemin Joseph Aiguier, 13402 Marseille Cedex 20, France
| | - Houcemeddine Othman
- Laboratoire des Venins et Biomolécules Thérapeutiques LR11IPT08, Université Tunis-El Manar, Institut Pasteur de Tunis , Tunis 1002, Tunisia
| | - Neila Achouri
- Laboratory of Biochemistry and Enzymatic Engineering of Lipases, ENIS , 3038 Sfax, Tunisia
| | - Alexandre Noiriel
- Univ Lyon, Université Lyon 1, Institut de Chimie et de Biochimie Moléculaires et Supramoléculaires (ICBMS), UMR 5246 CNRS, Métabolisme, Enzymes et Mécanismes Moléculaires (MEM2 , F-69622 Villeurbanne cedex, France
| | - Vincent Arondel
- Univ Bordeaux, UMR 5200, Laboratoire de Biogenèse Membranaire , Bat. A3 Campus INRA de Bordeaux 71 avenue E., Bourlaux CS 2003233140 Villenave d'Ornon, France
| | - Najet Srairi-Abid
- Laboratoire des Venins et Biomolécules Thérapeutiques LR11IPT08, Université Tunis-El Manar, Institut Pasteur de Tunis , Tunis 1002, Tunisia
| | - Abdelkarim Abousalham
- Univ Lyon, Université Lyon 1, Institut de Chimie et de Biochimie Moléculaires et Supramoléculaires (ICBMS), UMR 5246 CNRS, Métabolisme, Enzymes et Mécanismes Moléculaires (MEM2 , F-69622 Villeurbanne cedex, France
| | - Youssef Gargouri
- Laboratory of Biochemistry and Enzymatic Engineering of Lipases, ENIS , 3038 Sfax, Tunisia
| | - Nabil Miled
- Laboratory of Biochemistry and Enzymatic Engineering of Lipases, ENIS , 3038 Sfax, Tunisia
| | - Ahmed Fendri
- Laboratory of Biochemistry and Enzymatic Engineering of Lipases, ENIS , 3038 Sfax, Tunisia
| |
Collapse
|
38
|
Fernandes CAH, Pazin WM, Dreyer TR, Bicev RN, Cavalcante WLG, Fortes-Dias CL, Ito AS, Oliveira CLP, Fernandez RM, Fontes MRM. Biophysical studies suggest a new structural arrangement of crotoxin and provide insights into its toxic mechanism. Sci Rep 2017; 7:43885. [PMID: 28256632 PMCID: PMC5335569 DOI: 10.1038/srep43885] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 01/31/2017] [Indexed: 11/19/2022] Open
Abstract
Crotoxin (CTX) is the main neurotoxin found in Crotalus durissus rattlesnake venoms being composed by a nontoxic and non-enzymatic component (CA) and a toxic phospholipase A2 (CB). Previous crystallographic structures of CTX and CB provided relevant insights: (i) CTX structure showed a 1:1 molecular ratio between CA and CB, presenting three tryptophan residues in the CA/CB interface and one exposed to solvent; (ii) CB structure displayed a tetrameric conformation. This study aims to provide further information on the CTX mechanism of action by several biophysical methods. Our data show that isolated CB can in fact form tetramers in solution; however, these tetramers can be dissociated by CA titration. Furthermore, CTX exhibits a strong reduction in fluorescence intensity and lifetime compared with isolated CA and CB, suggesting that all tryptophan residues in CTX may be hidden by the CA/CB interface. By companying spectroscopy fluorescence and SAXS data, we obtained a new structural model for the CTX heterodimer in which all tryptophans are located in the interface, and the N-terminal region of CB is largely exposed to the solvent. Based on this model, we propose a toxic mechanism of action for CTX, involving the interaction of N-terminal region of CB with the target before CA dissociation.
Collapse
Affiliation(s)
- Carlos A. H. Fernandes
- Departamento de Física e Biofísica, Instituto de Biociências, Universidade Estadual Paulista, UNESP, Botucatu-SP, Brazil
| | - Wallance M. Pazin
- Departamento de Física, Faculdade de Filosofia Ciências e Letras de Ribeirão Preto, USP, Ribeirão Preto-SP, Brazil
| | - Thiago R. Dreyer
- Departamento de Física e Biofísica, Instituto de Biociências, Universidade Estadual Paulista, UNESP, Botucatu-SP, Brazil
| | - Renata N. Bicev
- Departamento de Física Experimental, Instituto de Física, Universidade de São Paulo – USP, São Paulo, SP, Brazil
| | - Walter L. G. Cavalcante
- Departamento de Física e Biofísica, Instituto de Biociências, Universidade Estadual Paulista, UNESP, Botucatu-SP, Brazil
- Departamento de Farmacologia, Instituto de Ciências Biológicas, UFMG, Belo Horizonte, MG, Brazil
| | - Consuelo L. Fortes-Dias
- Diretoria de Pesquisa e Desenvolvimento, Fundação Ezequiel Dias (FUNED), Belo Horizonte, MG, Brazil
| | - Amando S. Ito
- Departamento de Física, Faculdade de Filosofia Ciências e Letras de Ribeirão Preto, USP, Ribeirão Preto-SP, Brazil
| | - Cristiano L. P. Oliveira
- Departamento de Física Experimental, Instituto de Física, Universidade de São Paulo – USP, São Paulo, SP, Brazil
| | - Roberto Morato Fernandez
- Departamento de Física e Biofísica, Instituto de Biociências, Universidade Estadual Paulista, UNESP, Botucatu-SP, Brazil
| | - Marcos R. M. Fontes
- Departamento de Física e Biofísica, Instituto de Biociências, Universidade Estadual Paulista, UNESP, Botucatu-SP, Brazil
| |
Collapse
|
39
|
Nemati R, Dietz C, Anstadt E, Clark R, Smith M, Nichols F, Yao X. Simultaneous Determination of Absolute Configuration and Quantity of Lipopeptides Using Chiral Liquid Chromatography/Mass Spectrometry and Diastereomeric Internal Standards. Anal Chem 2017; 89:3583-3589. [DOI: 10.1021/acs.analchem.6b04901] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Reza Nemati
- Department
of Chemistry, University of Connecticut, Storrs, Connecticut 06269-3060, United States
| | - Christopher Dietz
- Department
of Chemistry, University of Connecticut, Storrs, Connecticut 06269-3060, United States
| | - Emily Anstadt
- Department
of Immunology and Medicine, University of Connecticut School of Medicine, Farmington, Connecticut 06030, United States
| | - Robert Clark
- Department
of Immunology and Medicine, University of Connecticut School of Medicine, Farmington, Connecticut 06030, United States
| | - Michael Smith
- Department
of Chemistry, University of Connecticut, Storrs, Connecticut 06269-3060, United States
| | - Frank Nichols
- Department
of Oral Health and Diagnostic Sciences, University of Connecticut School of Dental Medicine, Farmington, Connecticut 06030, United States
| | - Xudong Yao
- Department
of Chemistry, University of Connecticut, Storrs, Connecticut 06269-3060, United States
- Institute
for Systems Genomics, University of Connecticut, Storrs, Connecticut 06269, United States
| |
Collapse
|
40
|
Shang M, Xie Z, Tang Z, He L, Wang X, Wang C, Wu Y, Li Y, Zhao L, Lv Z, Wu Z, Huang Y, Yu X, Li X. Expression of Clonorchis sinensis GIIIsPLA 2 protein in baculovirus-infected insect cells and its overexpression facilitating epithelial-mesenchymal transition in Huh7 cells via AKT pathway. Parasitol Res 2017; 116:1307-1316. [PMID: 28220242 DOI: 10.1007/s00436-017-5409-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 02/12/2017] [Indexed: 12/29/2022]
Abstract
Although prior studies confirmed that group III secretory phospholipase A2 of Clonorchis sinensis (CsGIIIsPLA2) had stimulating effect on liver fibrosis by binding to LX-2 cells, large-scale expression of recombinant protein and its function in the progression of hepatoma are worth exploring. Because of high productivity and low lipopolysaccharides (LPS) in the Sf9-baculovirus expression system, we firstly used this system to express the coding region of CsGIIIsPLA2. The molecular weight of recombinant CsGIIIsPLA2 protein was about 34 kDa. Further investigation showed that most of the recombinant protein presented intracellular expression in Sf9 insect cell nucleus and could be detected only into cell debris, which made the protein purification and further functional study difficult. Therefore, to study the role of CsGIIIsPLA2 in hepatocellular carcinoma (HCC) progression, CsGIIIsPLA2 overexpression Huh7 cell model was applied. Cell proliferation, migration, and the expression level of epithelial-mesenchymal transition (EMT)-related molecules (E-cadherin, N-cadherin, α-catenin, Vimentin, p300, Snail, and Slug) along with possible mechanism were measured. The results indicated that CsGIIIsPLA2 overexpression not only inhibited cell proliferation and promoted migration and EMT but also enhanced the phosphorylation of AKT in HCC cells. In conclusion, this study supported that CsGIIIsPLA2 overexpression suppressed cell proliferation and induced EMT through the AKT pathway.
Collapse
Affiliation(s)
- Mei Shang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China.,Key Laboratory for Tropical Diseases Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China
| | - Zhizhi Xie
- Department of Clinical Laboratory, Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China
| | - Zeli Tang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China.,Key Laboratory for Tropical Diseases Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China
| | - Lei He
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China.,Key Laboratory for Tropical Diseases Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China.,Department of Clinical Laboratory, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, 510060, People's Republic of China
| | - Xiaoyun Wang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China.,Key Laboratory for Tropical Diseases Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China
| | - Caiqin Wang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China.,Key Laboratory for Tropical Diseases Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China
| | - Yinjuan Wu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China.,Key Laboratory for Tropical Diseases Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China
| | - Ye Li
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China.,Key Laboratory for Tropical Diseases Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China
| | - Lu Zhao
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China.,Key Laboratory for Tropical Diseases Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China
| | - Zhiyue Lv
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China.,Key Laboratory for Tropical Diseases Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China
| | - Zhongdao Wu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China.,Key Laboratory for Tropical Diseases Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China
| | - Yan Huang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China.,Key Laboratory for Tropical Diseases Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China
| | - Xinbing Yu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China.,Key Laboratory for Tropical Diseases Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China
| | - Xuerong Li
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China. .,Key Laboratory for Tropical Diseases Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China.
| |
Collapse
|
41
|
Zambelli VO, Chioato L, Gutierrez VP, Ward RJ, Cury Y. Structural determinants of the hyperalgesic activity of myotoxic Lys49-phospholipase A 2. J Venom Anim Toxins Incl Trop Dis 2017; 23:7. [PMID: 28203248 PMCID: PMC5303236 DOI: 10.1186/s40409-017-0099-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 02/01/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Bothropstoxin-I (BthTx-I) is a Lys49-phospholipase A2 (Lys49-PLA2) from the venom of Bothrops jararacussu, which despite of the lack of catalytic activity induces myotoxicity, inflammation and pain. The C-terminal region of the Lys49-PLA2s is important for these effects; however, the amino acid residues that determine hyperalgesia and edema are unknown. The aim of this study was to characterize the structural determinants for the Lys49-PLA2-induced nociception and inflammation. METHODS Scanning alanine mutagenesis in the active-site and C-terminal regions of BthTx-I has been used to study the structural determinants of toxin activities. The R118A mutant was employed as this substitution decreases PLA2 myotoxicity. In addition, K115A and K116A mutants - which contribute to decrease cytotoxicity - and the K122A mutant - which decreases both myotoxicity and cytotoxicity - were also used. The H48Q mutant - which does not interfere with membrane damage or myotoxic activity - was used to evaluate if the PLA2 catalytic site is relevant for the non-catalytic PLA2-induced pain and inflammation. Wistar male rats received intraplantar injections with mutant PLA2. Subsequently, hyperalgesia and edema were evaluated by the paw pressure test and by a plethysmometer. Native and recombinant BthTx-I were used as controls. RESULTS Native and recombinant BthTx-I induced hyperalgesia and edema, which peaked at 2 h. The R118A mutant did not induce nociception or edema. The mutations K115A and K116A abolished hyperalgesia without interfering with edema. Finally, the K122A mutant did not induce hyperalgesia and presented a decreased inflammatory response. CONCLUSIONS The results obtained with the BthTx-I mutants suggest, for the first time, that there are distinct residues responsible for the hyperalgesia and edema induced by BthTx-I. In addition, we also showed that cytolytic activity is essential for the hyperalgesic effect but not for edematogenic activity, corroborating previous data showing that edema and hyperalgesia can occur in a non-dependent manner. Understanding the structure-activity relationship in BthTx-I has opened new possibilities to discover the target for PLA2-induced pain.
Collapse
Affiliation(s)
- Vanessa Olzon Zambelli
- Butantan Institute, Special Laboratory for Pain and Signaling, Av. Vital Brazil, 1500, São Paulo, SP CEP 05503-900 Brazil
| | - Lucimara Chioato
- Department of Chemistry, School of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP Brazil
| | - Vanessa Pacciari Gutierrez
- Butantan Institute, Special Laboratory for Pain and Signaling, Av. Vital Brazil, 1500, São Paulo, SP CEP 05503-900 Brazil
| | - Richard John Ward
- Department of Chemistry, School of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP Brazil
| | - Yara Cury
- Butantan Institute, Special Laboratory for Pain and Signaling, Av. Vital Brazil, 1500, São Paulo, SP CEP 05503-900 Brazil
| |
Collapse
|
42
|
Babon A, Wurceldorf T, Almunia C, Pichard S, Chenal A, Buhot C, Beaumelle B, Gillet D. Bee venom phospholipase A2 as a membrane-binding vector for cell surface display or internalization of soluble proteins. Toxicon 2016; 116:56-62. [PMID: 26253725 DOI: 10.1016/j.toxicon.2015.07.338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 07/28/2015] [Accepted: 07/31/2015] [Indexed: 11/30/2022]
Abstract
We showed that bee venom phospholipase A2 can be used as a membrane-binding vector to anchor to the surface of cells a soluble protein fused to its C-terminus. ZZ, a two-domain derivative of staphylococcal protein A capable of binding constant regions of antibodies was fused to the C-terminus of the phospholipase or to a mutant devoid of enzymatic activity. The fusion proteins bound to the surface of cells and could themselves bind IgGs. Their fate depended on the cell type to which they bound. On the A431 carcinoma cell line the proteins remained exposed on the cell surface. In contrast, on human dendritic cells the proteins were internalized into early endosomes.
Collapse
Affiliation(s)
- Aurélie Babon
- CEA, iBiTecS, SIMOPRO, LabEx LERMIT, Gif sur Yvette F-91191, France
| | | | | | - Sylvain Pichard
- CEA, iBiTecS, SIMOPRO, LabEx LERMIT, Gif sur Yvette F-91191, France
| | - Alexandre Chenal
- CEA, iBiTecS, SIMOPRO, LabEx LERMIT, Gif sur Yvette F-91191, France
| | - Cécile Buhot
- CEA, iBiTecS, SIMOPRO, LabEx LERMIT, Gif sur Yvette F-91191, France
| | - Bruno Beaumelle
- FRE 3689 CNRS-UM, 1919 Route de Mende, 34293 Montpellier Cedex 05, France
| | - Daniel Gillet
- CEA, iBiTecS, SIMOPRO, LabEx LERMIT, Gif sur Yvette F-91191, France.
| |
Collapse
|
43
|
Matricardi PM, Kleine-Tebbe J, Hoffmann HJ, Valenta R, Hilger C, Hofmaier S, Aalberse RC, Agache I, Asero R, Ballmer-Weber B, Barber D, Beyer K, Biedermann T, Bilò MB, Blank S, Bohle B, Bosshard PP, Breiteneder H, Brough HA, Caraballo L, Caubet JC, Crameri R, Davies JM, Douladiris N, Ebisawa M, EIgenmann PA, Fernandez-Rivas M, Ferreira F, Gadermaier G, Glatz M, Hamilton RG, Hawranek T, Hellings P, Hoffmann-Sommergruber K, Jakob T, Jappe U, Jutel M, Kamath SD, Knol EF, Korosec P, Kuehn A, Lack G, Lopata AL, Mäkelä M, Morisset M, Niederberger V, Nowak-Węgrzyn AH, Papadopoulos NG, Pastorello EA, Pauli G, Platts-Mills T, Posa D, Poulsen LK, Raulf M, Sastre J, Scala E, Schmid JM, Schmid-Grendelmeier P, van Hage M, van Ree R, Vieths S, Weber R, Wickman M, Muraro A, Ollert M. EAACI Molecular Allergology User's Guide. Pediatr Allergy Immunol 2016; 27 Suppl 23:1-250. [PMID: 27288833 DOI: 10.1111/pai.12563] [Citation(s) in RCA: 535] [Impact Index Per Article: 59.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The availability of allergen molecules ('components') from several protein families has advanced our understanding of immunoglobulin E (IgE)-mediated responses and enabled 'component-resolved diagnosis' (CRD). The European Academy of Allergy and Clinical Immunology (EAACI) Molecular Allergology User's Guide (MAUG) provides comprehensive information on important allergens and describes the diagnostic options using CRD. Part A of the EAACI MAUG introduces allergen molecules, families, composition of extracts, databases, and diagnostic IgE, skin, and basophil tests. Singleplex and multiplex IgE assays with components improve both sensitivity for low-abundance allergens and analytical specificity; IgE to individual allergens can yield information on clinical risks and distinguish cross-reactivity from true primary sensitization. Part B discusses the clinical and molecular aspects of IgE-mediated allergies to foods (including nuts, seeds, legumes, fruits, vegetables, cereal grains, milk, egg, meat, fish, and shellfish), inhalants (pollen, mold spores, mites, and animal dander), and Hymenoptera venom. Diagnostic algorithms and short case histories provide useful information for the clinical workup of allergic individuals targeted for CRD. Part C covers protein families containing ubiquitous, highly cross-reactive panallergens from plant (lipid transfer proteins, polcalcins, PR-10, profilins) and animal sources (lipocalins, parvalbumins, serum albumins, tropomyosins) and explains their diagnostic and clinical utility. Part D lists 100 important allergen molecules. In conclusion, IgE-mediated reactions and allergic diseases, including allergic rhinoconjunctivitis, asthma, food reactions, and insect sting reactions, are discussed from a novel molecular perspective. The EAACI MAUG documents the rapid progression of molecular allergology from basic research to its integration into clinical practice, a quantum leap in the management of allergic patients.
Collapse
Affiliation(s)
- P M Matricardi
- Paediatric Pneumology and Immunology, Charitè Medical University, Berlin, Germany
| | - J Kleine-Tebbe
- Allergy & Asthma Center Westend, Outpatient Clinic Ackermann, Hanf, & Kleine-Tebbe, Berlin, Germany
| | - H J Hoffmann
- Department of Respiratory Diseases and Allergy, Institute of Clinical Medicine, Aarhus University and Aarhus University Hospital, Aarhus, Denmark
| | - R Valenta
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - C Hilger
- Department of Infection & Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - S Hofmaier
- Paediatric Pneumology and Immunology, Charitè Medical University, Berlin, Germany
| | - R C Aalberse
- Sanquin Research, Department of Immunopathology, Amsterdam, The Netherlands
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - I Agache
- Department of Allergy and Clinical Immunology, Faculty of Medicine, Transylvania University of Brasov, Brasov, Romania
| | - R Asero
- Ambulatorio di Allergologia, Clinica San Carlo, Paderno Dugnano, Italy
| | - B Ballmer-Weber
- Allergy Unit, Department of Dermatology, University Hospital Zürich, Zürich, Switzerland
| | - D Barber
- IMMA-School of Medicine, University CEU San Pablo, Madrid, Spain
| | - K Beyer
- Paediatric Pneumology and Immunology, Charitè Medical University, Berlin, Germany
| | - T Biedermann
- Department of Dermatology and Allergology, Technical University Munich, Munich, Germany
| | - M B Bilò
- Allergy Unit, Department of Internal Medicine, University Hospital Ospedali Riuniti di Ancona, Ancona, Italy
| | - S Blank
- Center of Allergy and Environment (ZAUM), Helmholtz Center Munich, Technical University of Munich, Munich, Germany
| | - B Bohle
- Division of Experimental Allergology, Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Vienna, Austria
| | - P P Bosshard
- Allergy Unit, Department of Dermatology, University Hospital Zürich, Zürich, Switzerland
| | - H Breiteneder
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - H A Brough
- Paediatric Allergy, Department of Asthma, Allergy and Respiratory Science, King's College London, Guys' Hospital, London, UK
| | - L Caraballo
- Institute for Immunological Research, The University of Cartagena, Cartagena de Indias, Colombia
| | - J C Caubet
- Pediatric Allergy Unit, Department of Child and Adolescent, University Hospitals of Geneva, Geneva, Switzerland
| | - R Crameri
- Swiss Institute of Allergy and Asthma Research, University of Zürich, Davos, Switzerland
| | - J M Davies
- School of Biomedical Sciences, Institute of Biomedical Innovation, Queensland University of Technology, Brisbane, Qld, Australia
| | - N Douladiris
- Allergy Unit, 2nd Paediatric Clinic, National & Kapodistrian University, Athens, Greece
| | - M Ebisawa
- Department of Allergy, Clinical Research Center for Allergology and Rheumatology, Sagamihara National Hospital, Kanagawa, Japan
| | - P A EIgenmann
- Pediatric Allergy Unit, Department of Child and Adolescent, University Hospitals of Geneva, Geneva, Switzerland
| | - M Fernandez-Rivas
- Allergy Department, Hospital Clinico San Carlos IdISSC, Madrid, Spain
| | - F Ferreira
- Division of Allergy and Immunology, Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - G Gadermaier
- Division of Allergy and Immunology, Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - M Glatz
- Allergy Unit, Department of Dermatology, University Hospital Zürich, Zürich, Switzerland
- Christine Kühne Center for Allergy Research and Education CK-CARE, Davos, Switzerland
| | - R G Hamilton
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - T Hawranek
- Department of Dermatology, Paracelsus Private Medical University, Salzburg, Austria
| | - P Hellings
- Department of Otorhinolaryngology, Academic Medical Center (AMC), Amsterdam, The Netherlands
- Department of Otorhinolaryngology, University Hospitals Leuven, Leuven, Belgium
| | - K Hoffmann-Sommergruber
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - T Jakob
- Department of Dermatology and Allergology, University Medical Center Giessen and Marburg, Justus Liebig University Giessen, Giessen, Germany
| | - U Jappe
- Division of Clinical and Molecular Allergology, Research Centre Borstel, Airway Research Centre North (ARCN), Member of the German Centre for Lung Research (DZL), Borstel, Germany
- Interdisciplinary Allergy Division, Department of Pneumology, University of Lübeck, Lübeck, Germany
| | - M Jutel
- Department of Clinical Immunology, 'ALL-MED' Medical Research Institute, Wrocław Medical University, Wrocław, Poland
| | - S D Kamath
- Molecular Allergy Research Laboratory, Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Townsville City, Qld, Australia
| | - E F Knol
- Departments of Immunology and Dermatology/Allergology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - P Korosec
- University Clinic of Respiratory and Allergic Diseases, Golnik, Slovenia
| | - A Kuehn
- Department of Infection & Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - G Lack
- King's College London, MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
- Division of Asthma, Allergy and Lung Biology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - A L Lopata
- Department of Clinical Immunology, 'ALL-MED' Medical Research Institute, Wrocław Medical University, Wrocław, Poland
| | - M Mäkelä
- Skin and Allergy Hospital, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - M Morisset
- National Service of Immuno-Allergology, Centre Hospitalier Luxembourg (CHL), Luxembourg, UK
| | - V Niederberger
- Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| | - A H Nowak-Węgrzyn
- Pediatric Allergy and Immunology, Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - N G Papadopoulos
- Centre for Paediatrics and Child Health, Institute of Human Development, University of Manchester, Manchester, UK
| | - E A Pastorello
- Unit of Allergology and Immunology, Niguarda Ca' Granda Hospital, Milan, Italy
| | - G Pauli
- Service de Pneumologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - T Platts-Mills
- Department of Microbiology & Immunology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - D Posa
- Paediatric Pneumology and Immunology, Charitè Medical University, Berlin, Germany
| | - L K Poulsen
- Allergy Clinic, Copenhagen University Hospital, Copenhagen, Denmark
| | - M Raulf
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Ruhr-University Bochum (IPA), Bochum, Germany
| | - J Sastre
- Allergy Division, Fundación Jimenez Díaz, Madrid, Spain
| | - E Scala
- Experimental Allergy Unit, IDI-IRCCS, Rome, Italy
| | - J M Schmid
- Department of Respiratory Diseases and Allergy, Institute of Clinical Medicine, Aarhus University and Aarhus University Hospital, Aarhus, Denmark
| | - P Schmid-Grendelmeier
- Allergy Unit, Department of Dermatology, University Hospital Zürich, Zürich, Switzerland
- Christine Kühne Center for Allergy Research and Education CK-CARE, Davos, Switzerland
| | - M van Hage
- Department of Medicine Solna, Clinical Immunology and Allergy Unit, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - R van Ree
- Departments of Experimental Immunology and of Otorhinolaryngology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - S Vieths
- Department of Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | - R Weber
- School of Medicine, University of Colorado, Denver, CO, USA
- Department of Medicine, National Jewish Health Service, Denver, CO, USA
| | - M Wickman
- Sachs' Children's Hospital, Karolinska Institutet, Stockholm, Sweden
| | - A Muraro
- The Referral Centre for Food Allergy Diagnosis and Treatment Veneto Region, Department of Mother and Child Health, University of Padua, Padua, Italy
| | - M Ollert
- Department of Infection & Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Department of Dermatology and Allergy Center, Odense Research Center for Anaphylaxis, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
44
|
Suladze S, Cinar S, Sperlich B, Winter R. Pressure Modulation of the Enzymatic Activity of Phospholipase A2, A Putative Membrane-Associated Pressure Sensor. J Am Chem Soc 2015; 137:12588-96. [DOI: 10.1021/jacs.5b07009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Saba Suladze
- Department of Chemistry and
Chemical Biology, Biophysical Chemistry, TU Dortmund University, Otto-Hahn-Str. 6, D-44221 Dortmund, Germany
| | - Suleyman Cinar
- Department of Chemistry and
Chemical Biology, Biophysical Chemistry, TU Dortmund University, Otto-Hahn-Str. 6, D-44221 Dortmund, Germany
| | - Benjamin Sperlich
- Department of Chemistry and
Chemical Biology, Biophysical Chemistry, TU Dortmund University, Otto-Hahn-Str. 6, D-44221 Dortmund, Germany
| | - Roland Winter
- Department of Chemistry and
Chemical Biology, Biophysical Chemistry, TU Dortmund University, Otto-Hahn-Str. 6, D-44221 Dortmund, Germany
| |
Collapse
|
45
|
Jridi I, Catacchio I, Majdoub H, Shahbazeddah D, El Ayeb M, Frassanito MA, Ribatti D, Vacca A, Borchani L. Hemilipin, a novel Hemiscorpius lepturus venom heterodimeric phospholipase A2, which inhibits angiogenesis in vitro and in vivo. Toxicon 2015; 105:34-44. [PMID: 26335363 DOI: 10.1016/j.toxicon.2015.08.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 08/14/2015] [Accepted: 08/26/2015] [Indexed: 11/30/2022]
Abstract
Phospholipases A2 (PLA2) are enzymes which specifically hydrolyze the sn-2 acyl ester bond of phospholipids producing free fatty acids and lysophospholipids. The secreted PLA2 (sPLA2) are the most common types of PLA2 purified from the snake venom, mammalian pancreatic juice and other sources. They display a variety of toxic actions and biological activities, including antitumoral and antiangiogenic effects. In this study, we report the isolation, characterization and the antiangiogenic activity of Hemilipin, a novel sPLA2 extracted from Hemiscorpius lepturus venom, the most dangerous scorpion in Iran. Hemilipin was purified by HPLC and analyzed by MALDI TOF/MS. The primary structure was determined by EDMAN degradation method and the PLA2 activity by titration of fatty acids released from the egg yolk phospholipids. Its antiangiogenic activity was studied in vitro by evaluating effects on apoptosis, Matrigel angiogenesis, migration and adhesion of human umbilical vein endothelial cells (HUVECs) and human pulmonary artery endothelial cells (HPAECs) and in vivo by the chorioallantoic membrane (CAM) assay. Mass spectrometry profile showed that Hemilipin is heterodimeric and the PLA2 test demonstrated its strong hydrolytic activity. N-terminal aminoacid sequence highlighted a significant homology of Hemilipin's small and large subunits with other sPLA2 group III. Hemilipin had no effect on apoptosis, but strongly impacted angiogenesis both in vitro and in vivo. Our results demonstrate that this novel non toxic sPLA2 could be a new tool to disrupt at different steps human angiogenesis.
Collapse
Affiliation(s)
- Imen Jridi
- Carthage University, Sciences Faculty of Bizerte, 7021 Jarzouna, Bizerte, Tunisia; Laboratory of Venom and Therapeutic Biomolecules, Pasteur Institute of Tunis, 13 Place Pasteur, BP 74, Tunis, 1002, Tunisia
| | - Ivana Catacchio
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari Medical School, Piazza Giulio Cesare 11, I-70124, Bari, Italy
| | - Hafed Majdoub
- USCR Protein Sequencer, Faculty of Sciences, Sfax, Tunisia
| | - Delavar Shahbazeddah
- Biotechnology Research Center, Medical Biotechnology Department, Venomics Lab, P.O.Box 13164943551 Tehran, Iran
| | - Mohamed El Ayeb
- Laboratory of Venom and Therapeutic Biomolecules, Pasteur Institute of Tunis, 13 Place Pasteur, BP 74, Tunis, 1002, Tunisia
| | - Maria Antonia Frassanito
- Department of Biomedical Sciences and Human Oncology, Section of Clinical Pathology, University of Bari Medical School, Piazza Giulio Cesare 11, I-70124, Bari, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs Section of Human Anatomy and Histology, University of Bari Medical School, National Cancer Institute "Giovanni Paolo II", Bari, Italy
| | - Angelo Vacca
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari Medical School, Piazza Giulio Cesare 11, I-70124, Bari, Italy
| | - Lamia Borchani
- Laboratory of Venom and Therapeutic Biomolecules, Pasteur Institute of Tunis, 13 Place Pasteur, BP 74, Tunis, 1002, Tunisia.
| |
Collapse
|
46
|
Damodaran S. Beyond the hydrophobic effect: Critical function of water at biological phase boundaries--A hypothesis. Adv Colloid Interface Sci 2015; 221:22-33. [PMID: 25888225 DOI: 10.1016/j.cis.2015.03.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 03/24/2015] [Accepted: 03/25/2015] [Indexed: 11/29/2022]
Abstract
Many life-sustaining processes in living cells occur at the membrane-water interface. The pertinent questions that need to be asked are what is the evolutionary reason for biology to choose the membrane-water interface as the site for performing and/or controlling crucial biological reactions and what is the key physical principle that is singular to the membrane-water interface that biology exploits for regulating metabolic processes in cells? In this review, a hypothesis is developed, which espouses that cells control activities of membrane-bound enzymes and receptor activated processes via manipulating the thermodynamic activity of water at the membrane-water interfacial region. In support of this hypothesis, first we establish that the surface pressure of a lipid monolayer is a direct measure of a reduction in the thermodynamic activity of interfacial water. Second, we show that the surface pressure-dependent activation/inactivation of interfacial enzymes is fundamentally related to their dependence on interfacial water activity. We extend this argument to infer that cells might manipulate activities of membrane-associated biological processes via manipulating the activity of interfacial water via localized compression or expansion of the interface. In this paper, we critically analyze literature data on mechano-activation of large pore ion channels in Escherichia coli spheroplasts and G-proteins in reconstituted lipid vesicles, and show that these pressure-induced activation processes are fundamentally and quantitatively related to changes in the thermodynamic state of interfacial water, caused by mechanical stretching of the bilayer.
Collapse
Affiliation(s)
- Srinivasan Damodaran
- University of Wisconsin-Madison, Department of Food Science, 1605 Linden Drive, Madison, WI 53706, United States.
| |
Collapse
|
47
|
Bp-13 PLA2: Purification and Neuromuscular Activity of a New Asp49 Toxin Isolated from Bothrops pauloensis Snake Venom. Biochem Res Int 2015; 2015:826059. [PMID: 25789175 PMCID: PMC4350628 DOI: 10.1155/2015/826059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 12/23/2014] [Accepted: 01/02/2015] [Indexed: 11/18/2022] Open
Abstract
A new PLA2 (Bp-13) was purified from Bothrops pauloensis snake venom after a single chromatographic step of RP-HPLC on μ-Bondapak C-18. Amino acid analysis showed a high content of hydrophobic and basic amino acids and 14 half-cysteine residues. The N-terminal sequence showed a high degree of homology with basic Asp49 PLA2 myotoxins from other Bothrops venoms. Bp-13 showed allosteric enzymatic behavior and maximal activity at pH 8.1, 36°–45°C. Full Bp-13 PLA2 activity required Ca2+; its PLA2 activity was inhibited by Mg2+, Mn2+, Sr2+, and Cd2+ in the presence and absence of 1 mM Ca2+. In the mouse phrenic nerve-diaphragm (PND) preparation, the time for 50% paralysis was concentration-dependent (P < 0.05). Both the replacement of Ca2+ by Sr2+ and temperature lowering (24°C) inhibited the Bp-13 PLA2-induced twitch-tension blockade. Bp-13 PLA2 inhibited the contractile response to direct electrical stimulation in curarized mouse PND preparation corroborating its contracture effect. In biventer cervicis preparations, Bp-13 induced irreversible twitch-tension blockade and the KCl evoked contracture was partially, but significantly, inhibited (P > 0.05). The main effect of this new Asp49 PLA2 of Bothrops pauloensis venom is on muscle fiber sarcolemma, with avian preparation being less responsive than rodent preparation. The study enhances biochemical and pharmacological characterization of B. pauloensis venom.
Collapse
|
48
|
Batchu KC, Hokynar K, Jeltsch M, Mattonet K, Somerharju P. Substrate efflux propensity is the key determinant of Ca2+-independent phospholipase A-β (iPLAβ)-mediated glycerophospholipid hydrolysis. J Biol Chem 2015; 290:10093-103. [PMID: 25713085 DOI: 10.1074/jbc.m115.642835] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Indexed: 12/13/2022] Open
Abstract
The A-type phospholipases (PLAs) are key players in glycerophospholipid (GPL) homeostasis and in mammalian cells; Ca(2+)-independent PLA-β (iPLAβ) in particular has been implicated in this essential process. However, the regulation of this enzyme, which is necessary to avoid futile competition between synthesis and degradation, is not understood. Recently, we provided evidence that the efflux of the substrate molecules from the bilayer is the rate-limiting step in the hydrolysis of GPLs by some secretory (nonhomeostatic) PLAs. To study whether this is the case with iPLAβ as well, a mass spectrometric assay was employed to determine the rate of hydrolysis of multiple saturated and unsaturated GPL species in parallel using micelles or vesicle bilayers as the macrosubstrate. With micelles, the hydrolysis decreased with increasing acyl chain length independent of unsaturation, and modest discrimination between acyl positional isomers was observed, presumably due to the differences in the structure of the sn-1 and sn-2 acyl-binding sites of the protein. In striking contrast, no significant discrimination between positional isomers was observed with bilayers, and the rate of hydrolysis decreased with the acyl chain length logarithmically and far more than with micelles. These data provide compelling evidence that efflux of the substrate molecule from the bilayer, which also decreases monotonously with acyl chain length, is the rate-determining step in iPLAβ-mediated hydrolysis of GPLs in membranes. This finding is intriguing as it may help to understand how homeostatic PLAs are regulated and how degradation and biosynthesis are coordinated.
Collapse
Affiliation(s)
| | - Kati Hokynar
- From the Departments of Biochemistry and Developmental Biology and
| | - Michael Jeltsch
- Biomedicine, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
| | - Kenny Mattonet
- Biomedicine, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
| | | |
Collapse
|
49
|
Water at Biological Phase Boundaries: Its Role in Interfacial Activation of Enzymes and Metabolic Pathways. Subcell Biochem 2015; 71:233-61. [PMID: 26438268 DOI: 10.1007/978-3-319-19060-0_10] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Many life-sustaining activities in living cells occur at the membrane-water interface. The pertinent questions that we need to ask are, what are the evolutionary reasons in biology for choosing the membrane-water interface as the site for performing and/or controlling crucial biological reactions, and what is the key physical principle that is very singular to the membrane-water interface that biology exploits for regulating metabolic processes in cells? In this chapter, a hypothesis is developed, which espouses that cells control activities of membrane-bound enzymes through manipulation of the thermodynamic activity of water in the lipid-water interfacial region. The hypothesis is based on the fact that the surface pressure of a lipid monolayer is a direct measure of the thermodynamic activity of water at the lipid-water interface. Accordingly, the surface pressure-dependent activation or inactivation of interfacial enzymes is directly related to changes in the thermodynamic activity of interfacial water. Extension of this argument suggests that cells may manipulate conformations (and activities) of membrane-bound enzymes by manipulating the (re)activity of interfacial water at various locations in the membrane by localized compression or expansion of the interface. In this respect, cells may use the membrane-bound hormone receptors, lipid phase transition, and local variations in membrane lipid composition as effectors of local compression and/or expansion of membrane, and thereby local water activity. Several experimental data in the literature will be reexamined in the light of this hypothesis.
Collapse
|
50
|
Clark J, Kay RR, Kielkowska A, Niewczas I, Fets L, Oxley D, Stephens LR, Hawkins PT. Dictyostelium uses ether-linked inositol phospholipids for intracellular signalling. EMBO J 2014; 33:2188-200. [PMID: 25180230 DOI: 10.15252/embj.201488677] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Inositol phospholipids are critical regulators of membrane biology throughout eukaryotes. The general principle by which they perform these roles is conserved across species and involves binding of differentially phosphorylated inositol head groups to specific protein domains. This interaction serves to both recruit and regulate the activity of several different classes of protein which act on membrane surfaces. In mammalian cells, these phosphorylated inositol head groups are predominantly borne by a C38:4 diacylglycerol backbone. We show here that the inositol phospholipids of Dictyostelium are different, being highly enriched in an unusual C34:1e lipid backbone, 1-hexadecyl-2-(11Z-octadecenoyl)-sn-glycero-3-phospho-(1'-myo-inositol), in which the sn-1 position contains an ether-linked C16:0 chain; they are thus plasmanylinositols. These plasmanylinositols respond acutely to stimulation of cells with chemoattractants, and their levels are regulated by PIPKs, PI3Ks and PTEN. In mammals and now in Dictyostelium, the hydrocarbon chains of inositol phospholipids are a highly selected subset of those available to other phospholipids, suggesting that different molecular selectors are at play in these organisms but serve a common, evolutionarily conserved purpose.
Collapse
Affiliation(s)
- Jonathan Clark
- Babraham Biosciences Technology Babraham Research Campus, Cambridge, UK
| | - Robert R Kay
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Anna Kielkowska
- Babraham Biosciences Technology Babraham Research Campus, Cambridge, UK
| | - Izabella Niewczas
- Babraham Biosciences Technology Babraham Research Campus, Cambridge, UK
| | - Louise Fets
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - David Oxley
- Signalling Programme Babraham Research Campus, Cambridge, UK
| | - Len R Stephens
- Signalling Programme Babraham Research Campus, Cambridge, UK
| | | |
Collapse
|