1
|
Chinigò G, Ruffinatti FA, Munaron L. The potential of TRP channels as new prognostic and therapeutic targets against prostate cancer progression. Biochim Biophys Acta Rev Cancer 2024; 1879:189226. [PMID: 39586480 DOI: 10.1016/j.bbcan.2024.189226] [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: 03/25/2024] [Revised: 10/28/2024] [Accepted: 11/21/2024] [Indexed: 11/27/2024]
Abstract
Prostate cancer (PCa) is the second deadliest cancer among men worldwide. Particularly critical is its development towards metastatic androgen-independent forms for which the current therapies are ineffective. Indeed, the 5-year relative survival for PCa drops dramatically to 34 % in the presence of metastases. The superfamily of Transient Receptor Potential (TRP) channels could answer the urgent request to identify new prognostic and therapeutic tools against metastatic PCa. Indeed, this class of ion channels revealed an appealing de-regulation during PCa development and its progression towards aggressive forms. Altered expression and/or functionality of several TRPs have been associated with the PCa metastatic cascade by significantly impacting tumor growth, invasiveness, and angiogenesis. In this review, we will dissect the contribution of TRP channels in such hallmarks of PCa and then discuss their applicability as new prognostic and therapeutic agents in the fight against metastatic PCa. In particular, the great potential of TRPM8, TRPV6, and TRPA1 in opening the way to new treatment perspectives will be highlighted.
Collapse
Affiliation(s)
- Giorgia Chinigò
- University of Turin, Department of Life Sciences and Systems Biology, via Accademia Albertina 13, 10123 Turin, Italy.
| | | | - Luca Munaron
- University of Turin, Department of Life Sciences and Systems Biology, via Accademia Albertina 13, 10123 Turin, Italy.
| |
Collapse
|
2
|
Lan Y, Qiu X, Xu Y. Expression, Purification and Characterization of Recombinant Disintegrin from Gloydius Brevicaudus Venom in Escherichia Coli. Protein J 2024; 43:603-612. [PMID: 38734856 DOI: 10.1007/s10930-024-10198-w] [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] [Accepted: 04/07/2024] [Indexed: 05/13/2024]
Abstract
Disintegrins, a family of snake venom protein, which are capable of modulating the activity of integrins that play a fundamental role in the regulation of many physiological and pathological processes. The main purpose of this study is to obtain the recombinant disintegrin (r-DI) and evaluate its biological activity. In this study, we explored a high-level expression prokaryotic system and purification strategy for r-DI. Then, r-DI was treated to assay effects on cell growth, migration, and invasion. The affinity for the interactions of r-DI with integrin was determined using Surface plasmon resonance (SPR) analyses. The r-DI can be expressed in Escherichia coli and purified by one-step chromatography. The r-DI can inhibit B16F10 cells proliferation, migration, and invasion. Also, we found that r-DI could interact with the integrin αIIbβ3 (GPIIb/IIIa). The r-DI can be expressed, purified, characterized through functional assays, and can also maintain strong biological activities. Thus, this study showed potential therapeutic effects of r-DI for further functional and structural studies.
Collapse
Affiliation(s)
- Yinxiang Lan
- Department of Pharmacy, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, FuJian Medical University, Fuzhou, Fujian, China
| | - Xiuliang Qiu
- Department of Pharmacy, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Yunlu Xu
- The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China.
- Center of Translational Hematology of Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
3
|
Wu Z, Wang C, Li C, Xu N, Cao X, Chen S, Shi Y, He Y, Zhang P, Ji J. Integrated Computational Pipeline for the High-Throughput Discovery of Cell Adhesion Peptides. J Phys Chem Lett 2024; 15:3748-3756. [PMID: 38551401 DOI: 10.1021/acs.jpclett.4c00393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Cell adhesion peptides (CAPs) often play a critical role in tissue engineering research. However, the discovery of novel CAPs for diverse applications remains a challenging and time-intensive process. This study presents an efficient computational pipeline integrating sequence embeddings, binding predictors, and molecular dynamics simulations to expedite the discovery of new CAPs. A Pro2vec model, trained on vast CAP data sets, was built to identify RGD-similar tripeptide candidates. These candidates were further evaluated for their binding affinity with integrin receptors using the Mutabind2 machine learning model. Additionally, molecular dynamics simulations were applied to model receptor-peptide interactions and calculate their binding free energies, providing a quantitative assessment of the binding strength for further screening. The resulting peptide demonstrated performance comparable to that of RGD in endothelial cell adhesion and spreading experimental assays, validating the efficacy of the integrated computational pipeline.
Collapse
Affiliation(s)
- Zhiyu Wu
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Institute of Zhejiang University-Quzhou, Quzhou 324000, China
| | - Cong Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China
| | - Chen Li
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Institute of Zhejiang University-Quzhou, Quzhou 324000, China
| | - Nan Xu
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Institute of Zhejiang University-Quzhou, Quzhou 324000, China
| | - Xiaoyong Cao
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Institute of Zhejiang University-Quzhou, Quzhou 324000, China
| | - Shengfu Chen
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Yao Shi
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Zhejiang University, Hangzhou 310058, China
| | - Yi He
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Institute of Zhejiang University-Quzhou, Quzhou 324000, China
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Peng Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory of Transvascular Implantation Devices, Qidi Road 456, Hangzhou 310058, China
| | - Jian Ji
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory of Transvascular Implantation Devices, Qidi Road 456, Hangzhou 310058, China
| |
Collapse
|
4
|
Asega AF, Barros BCSC, Chaves AFA, Oliveira AK, Bertholim L, Kitano ES, Serrano SMT. Mouse skin peptidomic analysis of the hemorrhage induced by a snake venom metalloprotease. Amino Acids 2023; 55:1103-1119. [PMID: 37389729 DOI: 10.1007/s00726-023-03299-w] [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: 05/16/2023] [Accepted: 06/22/2023] [Indexed: 07/01/2023]
Abstract
Hemorrhage induced by snake venom metalloproteases (SVMPs) results from proteolysis, capillary disruption, and blood extravasation. HF3, a potent SVMP of Bothrops jararaca, induces hemorrhage at pmol doses in the mouse skin. To gain insight into the hemorrhagic process, the main goal of this study was to analyze changes in the skin peptidome generated by injection of HF3, using approaches of mass spectrometry-based untargeted peptidomics. The results revealed that the sets of peptides found in the control and HF3-treated skin samples were distinct and derived from the cleavage of different proteins. Peptide bond cleavage site identification in the HF3-treated skin showed compatibility with trypsin-like serine proteases and cathepsins, suggesting the activation of host proteinases. Acetylated peptides, which originated from the cleavage at positions in the N-terminal region of proteins in both samples, were identified for the first time in the mouse skin peptidome. The number of peptides acetylated at the residue after the first Met residue, mostly Ser and Ala, was higher than that of peptides acetylated at the initial Met. Proteins cleaved in the hemorrhagic skin participate in cholesterol metabolism, PPAR signaling, and in the complement and coagulation cascades, indicating the impairment of these biological processes. The peptidomic analysis also indicated the emergence of peptides with potential biological activities, including pheromone, cell penetrating, quorum sensing, defense, and cell-cell communication in the mouse skin. Interestingly, peptides generated in the hemorrhagic skin promoted the inhibition of collagen-induced platelet aggregation and could act synergistically in the local tissue damage induced by HF3.
Collapse
Affiliation(s)
- Amanda F Asega
- Laboratory of Applied Toxinology, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Instituto Butantan, Av. Vital Brasil 1500, São Paulo, 05503-000, Brazil
| | - Bianca C S C Barros
- Laboratory of Applied Toxinology, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Instituto Butantan, Av. Vital Brasil 1500, São Paulo, 05503-000, Brazil
| | - Alison F A Chaves
- Laboratory of Applied Toxinology, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Instituto Butantan, Av. Vital Brasil 1500, São Paulo, 05503-000, Brazil
| | - Ana K Oliveira
- Laboratory of Applied Toxinology, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Instituto Butantan, Av. Vital Brasil 1500, São Paulo, 05503-000, Brazil
| | - Luciana Bertholim
- Laboratory of Applied Toxinology, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Instituto Butantan, Av. Vital Brasil 1500, São Paulo, 05503-000, Brazil
| | - Eduardo S Kitano
- Laboratory of Applied Toxinology, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Instituto Butantan, Av. Vital Brasil 1500, São Paulo, 05503-000, Brazil
| | - Solange M T Serrano
- Laboratory of Applied Toxinology, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Instituto Butantan, Av. Vital Brasil 1500, São Paulo, 05503-000, Brazil.
| |
Collapse
|
5
|
David V, Wermelinger LS, Frattani FS, Lima AGF, Santos YFS, Mourão PADS, Almeida FCL, Kurtenbach E, Zingali RB. rJararacin, a recombinant disintegrin from Bothrops jararaca venom: Exploring its effects on hemostasis and thrombosis. Arch Biochem Biophys 2023; 738:109557. [PMID: 36878339 DOI: 10.1016/j.abb.2023.109557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/24/2023] [Accepted: 03/02/2023] [Indexed: 03/07/2023]
Abstract
Integrins are a family of heterodimeric transmembrane receptors which link the extracellular matrix to the cell cytoskeleton. These receptors play a role in many cellular processes: adhesion, proliferation, migration, apoptosis, and platelet aggregation, thus modulating a wide range of scenarios in health and disease. Therefore, integrins have been the target of new antithrombotic drugs. Disintegrins from snake venoms are recognized by the ability to modulate the activity of integrins, such as integrin αIIbβ3, a fundamental platelet glycoprotein, and αvβ3 expressed on tumor cells. For this reason, disintegrins are unique and potential tools for examining integrin-matrix interaction and the development of novel antithrombotic agents. The present study aims to obtain the recombinant form of jararacin and evaluate the secondary structure and its effects on hemostasis and thrombosis. rJararacin was expressed in the Pichia pastoris (P. pastoris) expression system and purified the recombinant protein with a yield of 40 mg/L of culture. The molecular mass (7722 Da) and internal sequence were confirmed by mass spectrometry. Structure and folding analysis were obtained by Circular Dichroism and 1H Nuclear Magnetic Resonance spectra. Disintegrin structure reveals properly folded with the presence of β-sheet structure. rJararacin significantly demonstrated inhibition of the adhesion of B16F10 cells and platelets to the fibronectin matrix under static conditions. rJararacin inhibited platelet aggregation induced by ADP (IC50 95 nM), collagen (IC50 57 nM), and thrombin (IC50 22 nM) in a dose-dependent manner. This disintegrin also inhibited 81% and 94% of the adhesion of platelets to fibrinogen and collagen under continuous flow, respectively. In addition, rjararacin efficaciously prevents platelet aggregation in vitro and ex vivo with rat platelets and thrombus occlusion at an effective dose (5 mg/kg). The data here provides evidence that rjararacin possesses the potential as an αIIbβ3 antagonist, capable of preventing arterial thrombosis.
Collapse
Affiliation(s)
- Victor David
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, CEP 21941-902, Brazil.
| | - Luciana Serrão Wermelinger
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, CEP 21941-170, Brazil.
| | - Flávia Serra Frattani
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, CEP 21941-170, Brazil.
| | - Antonio Gilclêr Ferreira Lima
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, CEP 21941-902, Brazil.
| | - Yasmyn Fernandes Silva Santos
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, CEP 21941-902, Brazil.
| | - Paulo Antônio de Souza Mourão
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, CEP 21941-902, Brazil.
| | - Fabio Ceneviva Lacerda Almeida
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, CEP 21941-902, Brazil.
| | - Eleonora Kurtenbach
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, CEP 21941-170, Brazil.
| | - Russolina Benedeta Zingali
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, CEP 21941-902, Brazil.
| |
Collapse
|
6
|
Tsai TS, Tsai IH. Full sequencing and comparison of five venom metalloproteases of Trimeresurus gracilis: The PI-enzyme is most similar to okinalysin but the PIII-enzyme is most similar to Crotalus venom enzymes. Toxicon 2023; 225:107053. [PMID: 36758773 DOI: 10.1016/j.toxicon.2023.107053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023]
Abstract
The cDNAs encoding the Zn+2-metalloproteases (SVMPs) of Trimeresurus gracilis (abbreviated as Tgc), a pitviper endemic to Taiwan, were cloned from venom glands and sequenced. The amino-acid sequences of five novel SVMPs, including one P-III, three P-II and one P-I class enzymes, were thus deduced and subjected to BLAST-analyses. The P-III enzyme (designated as Tgc-PIII) is structurally most similar to the PIII-SVMPs of New World pitvipers but not similar to the PIII-SVMP of Ovophis okinavensis. Sequence-similarity analysis of 22 homologous PIII-SVMPs reveal three major structural subtypes of the pitviper PIII-SVMPs, which possibly have different substrate specificities. In addition, Tgc-PIII and the PI-class SVMP (named Tgc-MP) were isolated from the venom and verified by mass spectrometry. All the three deduced sequences of PII-SVMPs (Tgc-PIIs) contain an abnormal Zn+2-binding-site in their catalytic-domain, and an identical "long-disintegrin" domain. The predicted 85-residues disintegrin, gracilisin, bears high similarities to some long-disintegrins of the New-World pitvipers and salmosin3. By BLAST search and comparison, Tgc-MP is 96% similar to okinalysin, the hemorrhagic PI-SVMP of O. okinavensis, rather than any other PI-SVMPs in the databanks. Our results confirm the fast evolution of Tgc-SVMPs as well as their structural similarities to different SVMP-classes of the New-World pitvipers and of O. okinavensis, respectively. The implications of our findings are discussed along with our previous sequence comparisons of venom phospholipases A2 and ten venom serine proteases of Tgc.
Collapse
Affiliation(s)
- Tein-Shun Tsai
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Inn-Ho Tsai
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
7
|
Zhu J, Li X, Gao W, Jing J. Integrin Targeting Enhances the Antimelanoma Effect of Annexin V in Mice. Int J Mol Sci 2023; 24:ijms24043859. [PMID: 36835282 PMCID: PMC9959236 DOI: 10.3390/ijms24043859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/07/2023] [Accepted: 02/11/2023] [Indexed: 02/17/2023] Open
Abstract
Malignant melanoma, an increasingly common form of skin cancer, is a major threat to public health, especially when the disease progresses past skin lesions to the stage of advanced metastasis. Targeted drug development is an effective strategy for the treatment of malignant melanoma. In this work, a new antimelanoma tumor peptide, the lebestatin-annexin V (designated LbtA5) fusion protein, was developed and synthesized by recombinant DNA techniques. As a control, annexin V (designated ANV) was also synthesized by the same method. The fusion protein combines annexin V, which specifically recognizes and binds phosphatidylserine, with the disintegrin lebestatin (lbt), a polypeptide that specifically recognizes and binds integrin α1β1. LbtA5 was successfully prepared with good stability and high purity while retaining the dual biological activity of ANV and lbt. MTT assays demonstrated that both ANV and LbtA5 could reduce the viability of melanoma B16F10 cells, but the activity of the fusion protein LbtA5 was superior to that of ANV. The tumor volume growth was slowed in a mouse xenograft model treated with ANV and LbtA5, and the inhibitory effect of high concentrations of LbtA5 was significantly better than that of the same dose of ANV and was comparable to that of DTIC, a drug used clinically for melanoma treatment. The hematoxylin and eosin (H&E) staining test showed that ANV and LbtA5 had antitumor effects, but LbtA5 showed a stronger ability to induce melanoma necrosis in mice. Immunohistochemical experiments further showed that ANV and LbtA5 may inhibit tumor growth by inhibiting angiogenesis in tumor tissue. Fluorescence labeling experiments showed that the fusion of ANV with lbt enhanced the targeting of LbtA5 to mouse melanoma tumor tissue, and the amount of target protein in tumor tissue was significantly increased. In conclusion, effective coupling of the integrin α1β1-specific recognition molecule lbt confers stronger biological antimelanoma effects of ANV, which may be achieved by the dual effects of effective inhibition of B16F10 melanoma cell viability and inhibition of tumor tissue angiogenesis. The present study describes a new potential strategy for the application of the promising recombinant fusion protein LbtA5 in the treatment of various cancers, including malignant melanoma.
Collapse
Affiliation(s)
- Jingyi Zhu
- Beijing Key Lab of Biotechnology and Genetic Engineering, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Xiangning Li
- College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Wenling Gao
- College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Jian Jing
- Beijing Key Lab of Biotechnology and Genetic Engineering, College of Life Sciences, Beijing Normal University, Beijing 100875, China
- Correspondence: ; Tel.: +86-010-58802065
| |
Collapse
|
8
|
Zhang M, Sun Y, Zhang Y, Wang Z, Wang ZY, Ming XY, Guo ZD. Lipopolysaccharide and lipoteichoic acid regulate the PI3K/AKT pathway through osteopontin/integrin β3 to promote malignant progression of non-small cell lung cancer. J Thorac Dis 2023; 15:168-185. [PMID: 36794132 PMCID: PMC9922606 DOI: 10.21037/jtd-22-1825] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 01/06/2023] [Indexed: 01/18/2023]
Abstract
Background Lung cancer (LC) is a malignancy with one of the highest mortality rates. Respiratory microbiota is considered to play a key role in the development of LC, but the molecular mechanisms are rarely studied. Methods We used lipopolysaccharide (LPS) and lipoteichoic acid (LTA) to study human lung cancer cell lines PC9 and H1299. The gene expression of CXC chemokine ligand (CXCL)1/6, interleukin (IL)-6, IL-8, and tumor necrosis factor (TNF)-α were analyzed by quantitative real-time polymerase chain reaction (qRT-PCR). The Cell-Counting Kit 8 (CCK-8) was used to analyze cell proliferation. Transwell assays were performed to analyze cell migration ability. Flow cytometry was used to observe cell apoptosis. Western blot and qRT-PCR were used to analyze the expression of secreted phosphoprotein 1 (SPP1), toll-like receptor (TLR)-2/4, and NLR family pyrin domain containing 3 (NLRP3) to determine the mechanism of LPS + LTA. We evaluated the effect of LPS + LTA on cisplatin sensibility by analyzing cell proliferation, apoptosis, and caspase-3/9 expression levels. We observed the proliferation activity, apoptosis, and migration ability of cells in which SPP1 had been transfected small interfering (si) negative control (NC) and integrin β3 siRNA. Then the mRNA expression level and protein expression of PI3K, AKT, and ERK were analyzed. Finally, the nude mouse tumor transplantation model was conducted to verify. Results We studied that in two cell lines, the expression level of inflammatory factors in LPS+LTA group was significantly higher than that in single treatment group (P<0.001). We explored LPS + LTA combined treatment group significantly increased the expression of NLRP3 and genes and proteins. LPS + LTA + Cisplatin group could significantly reduce the inhibitory effect of LPS on cell proliferation (P<0.001), reduce the apoptosis rate (P<0.001) and significantly reduce the expression levels of caspase-3/9 (P<0.001) compared with Cisplatin group. Finally, we verified that LPS and LTA could increase osteopontin (OPN)/integrin β3 expression and activate the PI3K/AKT pathway to promote malignant progression of LC in vitro studies. Conclusions This study provides a theoretical basis for further exploration of the influence of lung microbiota on NSCLC and the optimization of LC treatment in the future.
Collapse
Affiliation(s)
- Miao Zhang
- Department of Oncology, Shijiazhuang People’s Hospital, Shijiazhuang, China
| | - Yi Sun
- Department of Oncology, Shijiazhuang People’s Hospital, Shijiazhuang, China
| | - Yan Zhang
- Department of Oncology, Shijiazhuang People’s Hospital, Shijiazhuang, China
| | - Zhen Wang
- Department of Oncology, Shijiazhuang People’s Hospital, Shijiazhuang, China
| | - Zhao-Yi Wang
- Department of Oncology, Shijiazhuang People’s Hospital, Shijiazhuang, China
| | - Xi-Yue Ming
- Department of Oncology, Shijiazhuang People’s Hospital, Shijiazhuang, China
| | - Zhen-Dong Guo
- Beijing Goldstandard Medicine Independent Clinical Laboratory Co. Ltd., Beijing, China
| |
Collapse
|
9
|
Oliveira AL, Viegas MF, da Silva SL, Soares AM, Ramos MJ, Fernandes PA. The chemistry of snake venom and its medicinal potential. Nat Rev Chem 2022; 6:451-469. [PMID: 35702592 PMCID: PMC9185726 DOI: 10.1038/s41570-022-00393-7] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2022] [Indexed: 12/15/2022]
Abstract
The fascination and fear of snakes dates back to time immemorial, with the first scientific treatise on snakebite envenoming, the Brooklyn Medical Papyrus, dating from ancient Egypt. Owing to their lethality, snakes have often been associated with images of perfidy, treachery and death. However, snakes did not always have such negative connotations. The curative capacity of venom has been known since antiquity, also making the snake a symbol of pharmacy and medicine. Today, there is renewed interest in pursuing snake-venom-based therapies. This Review focuses on the chemistry of snake venom and the potential for venom to be exploited for medicinal purposes in the development of drugs. The mixture of toxins that constitute snake venom is examined, focusing on the molecular structure, chemical reactivity and target recognition of the most bioactive toxins, from which bioactive drugs might be developed. The design and working mechanisms of snake-venom-derived drugs are illustrated, and the strategies by which toxins are transformed into therapeutics are analysed. Finally, the challenges in realizing the immense curative potential of snake venom are discussed, and chemical strategies by which a plethora of new drugs could be derived from snake venom are proposed.
Collapse
Affiliation(s)
- Ana L. Oliveira
- Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
- LAQV/Requimte, University of Porto, Porto, Portugal
| | - Matilde F. Viegas
- Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
- LAQV/Requimte, University of Porto, Porto, Portugal
| | - Saulo L. da Silva
- Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
- LAQV/Requimte, University of Porto, Porto, Portugal
| | - Andreimar M. Soares
- Biotechnology Laboratory for Proteins and Bioactive Compounds from the Western Amazon, Oswaldo Cruz Foundation, National Institute of Epidemiology in the Western Amazon (INCT-EpiAmO), Porto Velho, Brazil
- Sao Lucas Universitary Center (UniSL), Porto Velho, Brazil
| | - Maria J. Ramos
- Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
- LAQV/Requimte, University of Porto, Porto, Portugal
| | - Pedro A. Fernandes
- Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
- LAQV/Requimte, University of Porto, Porto, Portugal
| |
Collapse
|
10
|
Charoenkwan P, Schaduangrat N, Lio' P, Moni MA, Manavalan B, Shoombuatong W. NEPTUNE: A novel computational approach for accurate and large-scale identification of tumor homing peptides. Comput Biol Med 2022; 148:105700. [PMID: 35715261 DOI: 10.1016/j.compbiomed.2022.105700] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/31/2022] [Accepted: 06/04/2022] [Indexed: 11/16/2022]
Abstract
Tumor homing peptides (THPs) play a crucial role in recognizing and specifically binding to cancer cells. Although experimental approaches can facilitate the precise identification of THPs, they are usually time-consuming, labor-intensive, and not cost-effective. However, computational approaches can identify THPs by utilizing sequence information alone, thus highlighting their great potential for large-scale identification of THPs. Herein, we propose NEPTUNE, a novel computational approach for the accurate and large-scale identification of THPs from sequence information. Specifically, we constructed variant baseline models from multiple feature encoding schemes coupled with six popular machine learning algorithms. Subsequently, we comprehensively assessed and investigated the effects of these baseline models on THP prediction. Finally, the probabilistic information generated by the optimal baseline models is fed into a support vector machine-based classifier to construct the final meta-predictor (NEPTUNE). Cross-validation and independent tests demonstrated that NEPTUNE achieved superior performance for THP prediction compared with its constituent baseline models and the existing methods. Moreover, we employed the powerful SHapley additive exPlanations method to improve the interpretation of NEPTUNE and elucidate the most important features for identifying THPs. Finally, we implemented an online web server using NEPTUNE, which is available at http://pmlabstack.pythonanywhere.com/NEPTUNE. NEPTUNE could be beneficial for the large-scale identification of unknown THP candidates for follow-up experimental validation.
Collapse
Affiliation(s)
- Phasit Charoenkwan
- Modern Management and Information Technology, College of Arts, Media and Technology, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nalini Schaduangrat
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Pietro Lio'
- Department of Computer Science and Technology, University of Cambridge, Cambridge, CB3 0FD, UK
| | - Mohammad Ali Moni
- Artificial Intelligence & Digital Health, School of Health and Rehabilitation Sciences, Faculty of Health and Behavioural Sciences, The University of Queensland St Lucia, QLD, 4072, Australia
| | - Balachandran Manavalan
- Computational Biology and Bioinformatics Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, 16419, Gyeonggi-do, Republic of Korea.
| | - Watshara Shoombuatong
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand.
| |
Collapse
|
11
|
Bitiscetin-3, a Novel C-Type Lectin-like Protein Cloned from the Venom Gland of the Viper Bitis arietans, Induces Platelet Agglutination and Inhibits Binding of Von Willebrand Factor to Collagen. Toxins (Basel) 2022; 14:toxins14040236. [PMID: 35448845 PMCID: PMC9024624 DOI: 10.3390/toxins14040236] [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: 03/03/2022] [Revised: 03/18/2022] [Accepted: 03/18/2022] [Indexed: 02/04/2023] Open
Abstract
Bitiscetin-1 (aka bitiscetin) and bitiscetin-2 are C-type lectin-like proteins purified from the venom of Bitis arietans (puff adder). They bind to von Willebrand factor (VWF) and—at least bitiscetin-1—induce platelet agglutination via enhancement of VWF binding to platelet glycoprotein Ib (GPIb). Bitiscetin-1 and -2 bind the VWF A1 and A3 domains, respectively. The A3 domain includes the major site of VWF for binding collagen, explaining why bitiscetin-2 blocks VWF-to-collagen binding. In the present study, sequences for a novel bitiscetin protein—bitiscetin-3—were identified in cDNA constructed from the B. arietans venom gland. The deduced amino acid sequences of bitiscetin-3 subunits α and β share 79 and 80% identity with those of bitiscetin-1, respectively. Expression vectors for bitiscetin-3α and -3β were co-transfected to 293T cells, producing the heterodimer protein recombinant bitiscetin-3 (rBit-3). Functionally, purified rBit-3 (1) induced platelet agglutination involving VWF and GPIb, (2) did not compete with bitiscetin-1 for binding to VWF, (3) blocked VWF-to-collagen binding, and (4) lost its platelet agglutination inducing ability in the presence of an anti-VWF monoclonal antibody that blocked VWF-to-collagen binding. These combined results suggest that bitiscetin-3 binds to the A3 domain, as does bitiscetin-2. Except for a small N-terminal fragment of a single subunit—which differs from that of both bitiscetin-3 subunits—the sequences of bitiscetin-2 have never been determined. Therefore, by identifying and analyzing bitiscetin-3, the present study is the first to present the full-length α- and β-subunit sequences and recombinant expression of a bitiscetin-family toxin that blocks the binding of VWF to collagen.
Collapse
|
12
|
Alberto-Silva C, Querobino SM, Melo-Silva CA, Costa MS, Franco Oliveira LV, Zamuner SR. Local envenomation caused by a bioactive peptide fraction of Bothrops jararaca snake venom induces leukocyte influx in the lung and changes in pulmonary mechanics. Toxicon 2022; 207:52-59. [PMID: 34999120 DOI: 10.1016/j.toxicon.2022.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 01/02/2022] [Accepted: 01/03/2022] [Indexed: 11/30/2022]
Abstract
The crude venom of the Bothrops jararaca snake (Bj-CV) is a complex mixture of biologically active proteins that includes a variety of peptides in the low molecular weight fraction (Bj-PF). We investigated how an intramuscular injection of Bj-CV (1.2 mg kg-1) and Bj-PF (0.24 mg kg-1) influenced lung mechanics and lung and muscle inflammation in male Swiss mice 15 min, 1, 6, and 24 h after inoculation. Pressure dissipation against lung resistive components (ΔP1) rose significantly from 1 to 24 h after Bj-CV and 6-24 h after Bj-PF inoculation. Both Bj-CV and Bj-PF increased the total pressure variation of the lung (ΔPtot) 24 h after injection. Lung static elastance increased significantly after injection in all time periods investigated by Bj-CV and from 6 to 24 h by Bj-PF. Lung static elastance increased significantly after injection in all time periods investigated by Bj-CV and from 6 to 24 h by Bj-PF. Furthermore, intramuscular inoculation of Bj-CV and Bj-PF resulted in an increase in muscle and pulmonary inflammation, as evidenced by an increase in leukocyte influx when compared to the control group. Finally, both Bj-CV and Bj-PF cause acute lung injury, as shown by pulmonary inflammation and decreased lung mechanics. Furthermore, the fact that Bj-PF produces mechanical alterations in the lungs and muscular inflammation implies that non-enzymatic compounds can cause inflammation.
Collapse
Affiliation(s)
- Carlos Alberto-Silva
- Natural and Humanities Sciences Center, Experimental Morphophysiology Laboratory Federal University of ABC (UFABC), São Bernardo do Campo, 09606-070, SP, Brazil.
| | - Samyr Machado Querobino
- Natural and Humanities Sciences Center, Experimental Morphophysiology Laboratory Federal University of ABC (UFABC), São Bernardo do Campo, 09606-070, SP, Brazil; Minas Gerais State University (UEMG), Health and Biological Sciences Nucleus, Passos, MG, Brazil
| | | | - Maricilia Silva Costa
- Instituto de Pesquisa e Desenvolvimento - IP&D, Universidade do Vale do Paraíba- UNIVAP, Av. Shishima Hifumi 2911, São José dos Campos, Urbanova, 12244-000, SP, Brazil
| | - Luis Vicente Franco Oliveira
- Pós Graduate Program in Human Movement and Rehabilitation, Evangelical University of Goiás - UniEvangélica, Av. Universitária Km 3,5, Anápolis, 75083-515, Goiás, Brazil
| | - Stella Regina Zamuner
- Postgraduate Program in Medicine, Universidade Nove de Julho - UNINOVE, R: Vergueiro, 235 - Bairro Liberdade, São Paulo, SP, 01504-000, Brazil
| |
Collapse
|
13
|
Charoenkwan P, Chiangjong W, Nantasenamat C, Moni MA, Lio’ P, Manavalan B, Shoombuatong W. SCMTHP: A New Approach for Identifying and Characterizing of Tumor-Homing Peptides Using Estimated Propensity Scores of Amino Acids. Pharmaceutics 2022; 14:122. [PMID: 35057016 PMCID: PMC8779003 DOI: 10.3390/pharmaceutics14010122] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/16/2021] [Accepted: 12/28/2021] [Indexed: 12/13/2022] Open
Abstract
Tumor-homing peptides (THPs) are small peptides that can recognize and bind cancer cells specifically. To gain a better understanding of THPs' functional mechanisms, the accurate identification and characterization of THPs is required. Although some computational methods for in silico THP identification have been proposed, a major drawback is their lack of model interpretability. In this study, we propose a new, simple and easily interpretable computational approach (called SCMTHP) for identifying and analyzing tumor-homing activities of peptides via the use of a scoring card method (SCM). To improve the predictability and interpretability of our predictor, we generated propensity scores of 20 amino acids as THPs. Finally, informative physicochemical properties were used for providing insights on characteristics giving rise to the bioactivity of THPs via the use of SCMTHP-derived propensity scores. Benchmarking experiments from independent test indicated that SCMTHP could achieve comparable performance to state-of-the-art method with accuracies of 0.827 and 0.798, respectively, when evaluated on two benchmark datasets consisting of Main and Small datasets. Furthermore, SCMTHP was found to outperform several well-known machine learning-based classifiers (e.g., decision tree, k-nearest neighbor, multi-layer perceptron, naive Bayes and partial least squares regression) as indicated by both 10-fold cross-validation and independent tests. Finally, the SCMTHP web server was established and made freely available online. SCMTHP is expected to be a useful tool for rapid and accurate identification of THPs and for providing better understanding on THP biophysical and biochemical properties.
Collapse
Affiliation(s)
- Phasit Charoenkwan
- Modern Management and Information Technology, College of Arts, Media and Technology, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Wararat Chiangjong
- Pediatric Translational Research Unit, Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
| | - Chanin Nantasenamat
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand;
| | - Mohammad Ali Moni
- Artificial Intelligence & Digital Health Data Science, School of Health and Rehabilitation Sciences, Faculty of Health and Behavioural Sciences, The University of Queensland, St Lucia, QLD 4072, Australia;
| | - Pietro Lio’
- Department of Computer Science and Technology, University of Cambridge, Cambridge CB3 0FD, UK;
| | | | - Watshara Shoombuatong
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand;
| |
Collapse
|
14
|
Vasconcelos AA, Estrada JC, David V, Wermelinger LS, Almeida FCL, Zingali RB. Structure-Function Relationship of the Disintegrin Family: Sequence Signature and Integrin Interaction. Front Mol Biosci 2021; 8:783301. [PMID: 34926583 PMCID: PMC8678471 DOI: 10.3389/fmolb.2021.783301] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 11/11/2021] [Indexed: 01/09/2023] Open
Abstract
Disintegrins are small cysteine-rich proteins found in a variety of snake venom. These proteins selectively modulate integrin function, heterodimeric receptors involved in cell-cell and cell-matrix interaction that are widely studied as therapeutic targets. Snake venom disintegrins emerged from the snake venom metalloproteinase and are classified according to the sequence size and number of disulfide bonds. Evolutive structure and function diversification of disintegrin family involves a stepwise decrease in the polypeptide chain, loss of cysteine residues, and selectivity. Since the structure elucidation of echistatin, the description of the structural properties of disintegrins has allowed the investigation of the mechanisms involved in integrin-cell-extracellular matrix interaction. This review provides an analysis of the structures of all family groups enabling the description of an expanded classification of the disintegrin family in seven groups. Each group presents a particular disulfide pattern and sequence signatures, facilitating the identification of new disintegrins. The classification was based on the disintegrin-like domain of the human metalloproteinase (ADAM-10). We also present the sequence and structural signatures important for disintegrin-integrin interaction, unveiling the relationship between the structure and function of these proteins.
Collapse
Affiliation(s)
- Ariana A Vasconcelos
- Instituto de Bioquímica Médica (IBqM) Leopoldo de Meis, Centro Nacional de Ressonância Magnética Nuclear, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Centro Nacional de Ressonância Magnética Nuclear (CNRMN), Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jorge C Estrada
- Laboratório de Hemostase e Venenos, Instituto de Bioquímica Médica (IBqM) Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Victor David
- Laboratório de Hemostase e Venenos, Instituto de Bioquímica Médica (IBqM) Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana S Wermelinger
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fabio C L Almeida
- Instituto de Bioquímica Médica (IBqM) Leopoldo de Meis, Centro Nacional de Ressonância Magnética Nuclear, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Centro Nacional de Ressonância Magnética Nuclear (CNRMN), Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Russolina B Zingali
- Laboratório de Hemostase e Venenos, Instituto de Bioquímica Médica (IBqM) Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
15
|
Snake Venom Components: Tools and Cures to Target Cardiovascular Diseases. Molecules 2021; 26:molecules26082223. [PMID: 33921462 PMCID: PMC8070158 DOI: 10.3390/molecules26082223] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 12/31/2022] Open
Abstract
Cardiovascular diseases (CVDs) are considered as a major cause of death worldwide. Therefore, identifying and developing therapeutic strategies to treat and reduce the prevalence of CVDs is a major medical challenge. Several drugs used for the treatment of CVDs, such as captopril, emerged from natural products, namely snake venoms. These venoms are complex mixtures of bioactive molecules, which, among other physiological networks, target the cardiovascular system, leading to them being considered in the development and design of new drugs. In this review, we describe some snake venom molecules targeting the cardiovascular system such as phospholipase A2 (PLA2), natriuretic peptides (NPs), bradykinin-potentiating peptides (BPPs), cysteine-rich secretory proteins (CRISPs), disintegrins, fibrinolytic enzymes, and three-finger toxins (3FTXs). In addition, their molecular targets, and mechanisms of action—vasorelaxation, inhibition of platelet aggregation, cardioprotective activities—are discussed. The dissection of their biological effects at the molecular scale give insights for the development of future snake venom-derived drugs.
Collapse
|
16
|
Akhtar B, Muhammad F, Sharif A, Anwar MI. Mechanistic insights of snake venom disintegrins in cancer treatment. Eur J Pharmacol 2021; 899:174022. [PMID: 33727054 DOI: 10.1016/j.ejphar.2021.174022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 02/26/2021] [Accepted: 03/10/2021] [Indexed: 01/27/2023]
Abstract
Snake venoms are a potential source of various enzymatic and non-enzymatic compounds with a defensive role for the host. Various peptides with significant medicinal properties have been isolated and characterized from these venoms. Few of these are FDA approved. They inhibit tumor cells adhesion, migration, angiogenesis and metastasis by inhibiting integrins on transmembrane cellular surfaces. This plays important role in delaying tumor growth, neovascularization and development. Tumor targeting and smaller size make them ideal candidates as novel therapeutic agents for cancer treatment. This review is based on sources of these disintegrins, their targeting modality, classification and underlying anti-cancer potential.
Collapse
Affiliation(s)
- Bushra Akhtar
- Department of Pharmacy, University of Agriculture, Faisalabad, Pakistan.
| | - Faqir Muhammad
- Institute of Physiology and Pharmacology, University of Agriculture, Faisalabad, Pakistan
| | - Ali Sharif
- Institute of Pharmacy, Faculty of Pharmaceutical and Allied Health Sciences, Lahore College for Women University, Lahore, Pakistan
| | - Muhammad Irfan Anwar
- Department of Pathobiology, Faculty of Veterinary Sciences, Bahauddin Zakariya University, Multan, Pakistan
| |
Collapse
|
17
|
Cañas CA, Castaño-Valencia S, Castro-Herrera F, Cañas F, Tobón GJ. Biomedical applications of snake venom: from basic science to autoimmunity and rheumatology. J Transl Autoimmun 2020; 4:100076. [PMID: 33385156 PMCID: PMC7772571 DOI: 10.1016/j.jtauto.2020.100076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 12/26/2022] Open
Abstract
Snake venoms have components with diverse biological actions that are extensively studied to identify elements that may be useful in biomedical sciences. In the field of autoimmunity and rheumatology, various findings useful for the study of diseases and potential drug development have been reported. The study of disintegrins, proteins that block the action of integrins, has been useful for the development of antiplatelet agents and principles for the development of immunosuppressants and antineoplastics. Several proteins in snake venoms act on the coagulation cascade, activating factors that have allowed the development of tests for the study of coagulation, including Russell's viper venom time, which is useful in the diagnosis of antiphospholipid syndrome. Neurotoxins with either pre- or postsynaptic effects have been used to study neurogenic synapses and neuromuscular plaques and the development of analgesics, muscle relaxants and drugs for neurodegenerative diseases. Various components act by inhibiting cells and proteins of the immune system, which will allow the development of anti-inflammatory and immunosuppressive drugs. This review summarizes the usefulness of the components of snake venoms in the fields of autoimmunity and rheumatology, which can serve as a basis for diverse translational research.
Collapse
Affiliation(s)
- Carlos A Cañas
- GIRAT: Grupo de Investigación en Reumatología, Autoimunidad y Medicina Traslacional, Fundación Valle Del Lili and Universidad Icesi, Cali, Colombia.,Fundación Valle Del Lili, Rheumatology Unit, Cra 98 No. 18 - 49, Cali, 760032, Colombia
| | - Santiago Castaño-Valencia
- Department of Physiological Sciences, Department of Health Sciences, Universidad Del Valle, Cali, Colombia
| | - Fernando Castro-Herrera
- Department of Physiological Sciences, Department of Health Sciences, Universidad Del Valle, Cali, Colombia
| | - Felipe Cañas
- Department of Cardiology, Clínica Medellín, Medellín, Colombia
| | - Gabriel J Tobón
- GIRAT: Grupo de Investigación en Reumatología, Autoimunidad y Medicina Traslacional, Fundación Valle Del Lili and Universidad Icesi, Cali, Colombia.,Fundación Valle Del Lili, Rheumatology Unit, Cra 98 No. 18 - 49, Cali, 760032, Colombia
| |
Collapse
|
18
|
Luan J, Lu Y, Gao S, Zhang L. A potential inhibitory role for integrin in the receptor targeting of SARS-CoV-2. J Infect 2020; 81:318-356. [PMID: 32283163 PMCID: PMC7151354 DOI: 10.1016/j.jinf.2020.03.046] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 03/21/2020] [Accepted: 03/26/2020] [Indexed: 01/22/2023]
Affiliation(s)
- Junwen Luan
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250062, Shandong, China
| | - Yue Lu
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250062, Shandong, China; School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, Jinan 250200, Shandong, China
| | - Shan Gao
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250062, Shandong, China
| | - Leiliang Zhang
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250062, Shandong, China.
| |
Collapse
|
19
|
Golubev IV, Gureev VV, Korokin MV, Zatolokina MA, Avdeeva EV, Gureeva AV, Rozhkov IS, Serdyuk EA, Soldatova VA. Preclinical study of innovative peptides mimicking the space structure of the α-helix B of erythropoietin. RESEARCH RESULTS IN PHARMACOLOGY 2020. [DOI: 10.3897/rrpharmacology.6.55385] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Introduction: The aim of this study was to examine the effectiveness of innovative peptides obtained by addition of polypeptide motifs with antiaggregation activity (Arg-Gly-Asp, Lys-Gly-Asp and Pro-Gly-Pro) to a peptide mimicking the space structure of the α-helix B of erythropoietin pHBSP (Pyr-Glu-Gln-Leu-Glu-Arg-Ala-Leu-Asn-Ser-Ser).
Materials and methods: The cytoprotective activity of innovative peptides mimicking the space structure of the α-helix B of erythropoietin at the doses of 5 μg/ml, 30 μg/ml and 50 μg/ml was studied on human endothelial cell culture in a simulated oxidative stress. An ADMA-like model of preeclampsia was simulated in the experiment. The study was conducted in 260 female Wistar rats, weighing 250–300 g.
Results and discussion: Innovative peptides mimicking the space structure of the α-helix B of erythropoietin retain their cytoprotective activity in a simulated oxidative stress in HUVEC cell culture at the doses of 5 μg/ml, 30 μg/ml, and 50 μg/ml. The compounds with laboratory codes P-αB1 and P-αB3 had the most pronounced cytoprotective activity. Administration of N-nitro-L-arginine-methyl ether to pregnant females for 7 days causes the morphofunctional changes similar to clinical changes in preeclampsia. The innovative peptide under laboratory code P-αB4 at the dose of 50 μg/kg mimicking the space structure of the α-helix B of erythropoietin shows the most pronounced protective properties.
Conclusion: Innovative peptides mimicking the space structure of the α-helix B of erythropoietin have a pronounced positive influence on the morphofunctional disorders in animals with ADMA-like preeclampsia.
Collapse
|
20
|
Improved Antithrombotic Activity and Diminished Bleeding Side Effect of a PEGylated α IIbβ 3 Antagonist, Disintegrin. Toxins (Basel) 2020; 12:toxins12070426. [PMID: 32605221 PMCID: PMC7404706 DOI: 10.3390/toxins12070426] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 01/28/2023] Open
Abstract
Polymer polyethylene glycol (PEG), or PEGylation of polypeptides improves protein drug stability by decreasing degradation and reducing renal clearance. To produce a pharmaceutical disintegrin derivative, the N-terminal PEGylation technique was used to modify the disintegrin derivative [KGDRR]trimucrin for favorable safety, pharmacokinetic profiles, and antithrombotic efficacy. We compared intact [KGDRR]trimucrin (RR) and PEGylated KGDRR (PEG-RR) by in vitro and in vivo systems for their antithrombotic activities. The activity of platelet aggregation inhibition and the bleeding tendency side effect were also investigated. PEG-RR exhibited optimal potency in inhibiting platelet aggregation of human/mouse platelet-rich plasma activated by collagen or ADP with a lower IC50 than the intact derivative RR. In the illumination-induced mesenteric venous thrombosis model, RR and PEG-RR efficaciously prevented occlusive thrombosis in a dose-dependent manner. In rotational thromboelastometry assay, PEG-RR did not induce hypocoagulation in human whole blood even given at a higher concentration (30 μg/mL), while RR slightly prolonged clotting time. However, RR and PEG-RR were not associated with severe thrombocytopenia or bleeding in FcγRIIa-transgenic mice at equally efficacious antithrombotic dosages. We also found the in vivo half-life of PEGylation was longer than RR (RR: 15.65 h vs. PEG-RR: 20.45 h). In conclusion, injectable PEG-RR with prolonged half-life and decreased bleeding risk is a safer anti-thrombotic agent for long-acting treatment of thrombus diseases.
Collapse
|
21
|
A Novel α IIbβ 3 Antagonist from Snake Venom Prevents Thrombosis without Causing Bleeding. Toxins (Basel) 2019; 12:toxins12010011. [PMID: 31877725 PMCID: PMC7020592 DOI: 10.3390/toxins12010011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/16/2019] [Accepted: 12/19/2019] [Indexed: 12/29/2022] Open
Abstract
Life-threatening thrombocytopenia and bleeding, common side effects of clinically available αIIbβ3 antagonists, are associated with the induction of ligand-induced integrin conformational changes and exposure of ligand-induced binding sites (LIBSs). To address this issue, we examined intrinsic mechanisms and structure–activity relationships of purified disintegrins, from Protobothrops flavoviridis venom (i.e., Trimeresurus flavoviridis), TFV-1 and TFV-3 with distinctly different pro-hemorrhagic tendencies. TFV-1 with a different αIIbβ3 binding epitope from that of TFV-3 and chimeric 7E3 Fab, i.e., Abciximab, decelerates αIIbβ3 ligation without causing a conformational change in αIIbβ3, as determined with the LIBS antibody, AP5, and the mimetic, drug-dependent antibody (DDAb), AP2, an inhibitory monoclonal antibody raised against αIIbβ3. Consistent with their different binding epitopes, a combination of TFV-1 and AP2 did not induce FcγRIIa-mediated activation of the ITAM–Syk–PLCγ2 pathway and platelet aggregation, in contrast to the clinical antithrombotics, abciximab, eptifibatide, and disintegrin TFV-3. Furthermore, TFV-1 selectively inhibits Gα13-mediated platelet aggregation without affecting talin-driven clot firmness, which is responsible for physiological hemostatic processes. At equally efficacious antithrombotic dosages, TFV-1 caused neither severe thrombocytopenia nor bleeding in FcγRIIa-transgenic mice. Likewise, it did not induce hypocoagulation in human whole blood in the rotational thromboelastometry (ROTEM) assay used in perioperative situations. In contrast, TFV-3 and eptifibatide exhibited all of these hemostatic effects. Thus, the αIIbβ3 antagonist, TFV-1, efficaciously prevents arterial thrombosis without adversely affecting hemostasis.
Collapse
|