Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
1.
Int J Med Mushrooms ; 26(6): 53-68, 2024.
Article in English | MEDLINE | ID: mdl-38801087

ABSTRACT

The purification of a fibrinolytic enzyme from the fruiting bodies of wild-growing medicinal mushroom, Pycnoporus coccineus was achieved through a two-step procedure, resulting in its homogeneity. This purification process yielded a significant 4.13-fold increase in specific activity and an 8.0% recovery rate. The molecular weight of P. coccineus fibrinolytic enzyme (PCFE) was estimated to be 23 kDa using sodium dodecyl sulfate-polyacrylamide gel electrophoresis analysis. PCFE demonstrated its optimal activity at a temperature of 40 °C and pH 8. Notably, the enzymatic activity was inhibited by the presence of zinc or copper metal ions, as well as serine protease inhibitors, such as phenylmethylsulfonyl fluoride and 4-amidinophenylmethanesulfonyl fluoride. PCFE exhibited remarkable specificity towards a synthetic chromogenic substrate for thrombin. The enzyme demonstrated the Michaelis-Menten constant (Km), maximal velocity (V ), and catalytic rate constant (Kcat) values of 3.01 mM, 0.33 mM min-1 µg-1, and 764.1 s-1, respectively. In vitro assays showed PCFE's ability to effectively degrade fibrin and blood clots. The enzyme induced alterations in the density and structural characteristics of fibrin clots. PCFE exhibited significant effects on various clotting parameters, including recalcification time, activated partial thromboplastin time, prothrombin time, serotonin secretion from thrombin-activated platelets, and thrombin-induced acute thromboembolism. These findings suggest that P. coccineus holds potential as an antithrombotic biomaterials and resources for cardiovascular research.


Subject(s)
Fibrinolytic Agents , Pycnoporus , Serine Proteases , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/isolation & purification , Fibrinolytic Agents/chemistry , Serine Proteases/isolation & purification , Serine Proteases/pharmacology , Serine Proteases/metabolism , Serine Proteases/chemistry , Animals , Pycnoporus/enzymology , Molecular Weight , Fruiting Bodies, Fungal/chemistry , Hydrogen-Ion Concentration , Temperature , Humans , Fibrin/metabolism , Fungal Proteins/isolation & purification , Fungal Proteins/chemistry , Fungal Proteins/pharmacology
2.
J Biochem Mol Toxicol ; 37(11): e23456, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37439684

ABSTRACT

We aim to study the inhibitory effect of alkaline serine protease (ASPNJ) on lymphocytic leukemia Jurkat cells and its related mechanism through examining the expression of membrane proteins or membrane-associated proteins. MTT assay and trypan blue staining were used to detect the inhibitory effect of ASPNJ on the proliferation and growth of Jurkat cells. Wright-Giemsa staining was used to observe the effect of ASPNJ on the morphology of Jurkat cells. The effect of ASPNJ on Jurkat cell apoptosis was detected by flow cytometry. Two-dimensional electrophoresis-mass spectrometry (2-DE-MS) was used to detect and identify the differentially expressed proteins of Jurkat cells treated with ASPNJ (4 µg/mL, 3 h), of which three were selected and verified by Western blot. ASPNJ significantly inhibited the proliferation of leukemia cells (Raji, U937, and Jurkat), caused obvious morphological changes, and induced apoptosis of Jurkat cells. ASPNJ also increased the sensitivity of Jurkat cells to vincristine (VCR). Seven differentially expressed proteins were obtained through 2DE-MS, of which Peroxiredoxin-6 (PRDX6), Calcium-binding protein (CHP1), and 40S ribosomal protein SA (RPSA) were validated. ASPNJ can cause significant toxic effects on Jurkat cells and enhance the effects of VCR. The mechanism of action of ASPNJ on Jurkat cells may be related to differentially expressed proteins such as PRDX6. This study provides a new experimental basis and direction for antileukemia research.


Subject(s)
Serine Proteases , Serine , Humans , Jurkat Cells , Serine Proteases/pharmacology , Membrane Proteins , Cell Proliferation , Vincristine/pharmacology , Apoptosis , Serine Endopeptidases
3.
Respir Res ; 24(1): 133, 2023 May 17.
Article in English | MEDLINE | ID: mdl-37198686

ABSTRACT

BACKGROUND: Brensocatib is an oral, selective, reversible inhibitor of dipeptidyl peptidase-1 (DPP-1), responsible for activating neutrophil serine proteases (NSPs) including neutrophil elastase (NE), proteinase 3 (PR3), and cathepsin G (CatG). In chronic inflammatory lung diseases such as non-cystic fibrosis bronchiectasis (NCFBE), neutrophils accumulate in the airways resulting in excess active NSPs that cause damaging inflammation and lung destruction. METHODS: The 24-week WILLOW trial (NCT03218917) was a randomized, double-blind, placebo-controlled, parallel-group trial in patients with NCFBE conducted at 116 sites across 14 countries. In this trial, treatment with brensocatib was associated with improvements in clinical outcomes including time to first exacerbation, reduction in exacerbation frequency and a reduction in NE activity in sputum. An exploratory analysis of NE activity in white blood cell (WBC) extracts and NE, PR3 and CatG activity in sputum was conducted to further characterize brensocatib's effect and identify potential correlated effects. RESULTS: NE, PR3 and CatG activities were reduced in sputum and NE activity was reduced in WBC extracts in a dose-dependent manner after four weeks of brensocatib treatment, with a return to baseline four weeks after the end of treatment. Brensocatib produced the greatest reduction in the sputum activity of CatG, followed by NE and then PR3. Positive correlations among the sputum NSPs were observed both at baseline and in response to treatment, with the strongest correlation among the sputum NSPs for NE and CatG. CONCLUSIONS: These results suggest a broad anti-inflammatory effect of brensocatib underlying its clinical efficacy observed in NCFBE patients. TRIAL REGISTRATION: The study was approved by the corresponding ethical review boards of all participating centers. The trial was approved by the Food and Drug Administration and registered at clinicaltrials.gov (NCT03218917) on July 17, 2017 and approved by the European Medicines Agency and registered at the European Union Clinical trials Register (EudraCT No. 2017-002533-32). An independent, external data and safety monitoring committee (comprising physicians with pulmonary expertise, a statistician experienced in the evaluation of clinical safety, and experts in periodontal disease and dermatology) reviewed all adverse events.


Subject(s)
Bronchiectasis , Cystic Fibrosis , Salix , Humans , Serine Proteases/pharmacology , Serine Proteases/therapeutic use , Neutrophils , Bronchiectasis/diagnosis , Bronchiectasis/drug therapy , Leukocyte Elastase , Myeloblastin , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/pharmacology , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/therapeutic use
4.
J Chem Inf Model ; 63(7): 2122-2132, 2023 04 10.
Article in English | MEDLINE | ID: mdl-36943246

ABSTRACT

Molecular mechanics play an important role in enzyme action and understanding the dynamics of loop motion is key for designing inhibitors of an enzyme, particularly targeting the allosteric sites. For the successful creation of new protease inhibitors targeting the dengue serine protease, our current investigation detailed the intricate structural dynamics of NS2B/NS3 dengue protease. This enzyme is one of the most essential enzymes in the life cycle of the dengue virus, which is responsible for the activation/processing of viral polyprotein, thus making it a potential target for drug discovery. We showed that the internal dynamics of two regions, fingers 1 and 2 (R24-G39 and L149-A164, respectively) adjacent to the active site triad of this protease, control the enzyme action. Each of these regions is composed of two antiparallel ß-strands connected by ß-turn/hairpin loops. The correlated bending and rocking motions in the two ß-turns on either side of the active site were found to modulate the activity of the enzyme to a large extent. With increasing concentration of cosolvent dimethyl sulfoxide, correlated motions in the finger 2 region get diminished and bending of finger 1 increases, which are also reflected in the loss of enzyme activity. Decreasing temperature and mutations in neighboring nonsubstrate binding residues show similar effects on loop motion and enzyme kinetics. Therefore, in vitro noninvasive perturbation of these motions by the solvent exchange as well as cold stress in combination with in silico molecular dynamics simulations established the importance of the two ß-turns in the functioning of dengue virus serotype 2 NS2B/NS3 serine protease.


Subject(s)
Dengue Virus , Dengue , Humans , Solvents , Dengue Virus/metabolism , Viral Nonstructural Proteins/chemistry , Protease Inhibitors/pharmacology , Protease Inhibitors/chemistry , Dengue/drug therapy , Serine Proteases/pharmacology
5.
Toxicon ; 221: 106979, 2023 Jan 01.
Article in English | MEDLINE | ID: mdl-36423674

ABSTRACT

Serine Proteinase Associated Disintegrin-1 (SPAD-1) is a low molecular mass (26 kDa) positively charged protein purified from Russell's viper venom (RVV) possessing cytotoxic activity on MCF7, human breast cancer cells. Primary sequence analysis of the protein confirms that it is a novel Snake Venom Serine Proteinase (SVSP) and a member of the trypsin family. SPAD-1 contains a conserved triad of Histidine (H), Aspartic acid(D) and Serine(S) residues at its active site for proteinase activity and also an adjacent histidine-glycine-aspartic acid (HGD) disintegrin-like motif. The serine proteinase and disintegrin parts are functionally active and independent. SPAD-1 showed proteolytic digestion of fibrinogen and fibronectin, but laminin digestion was below the detectable limit. Proteolytically inactivated SPAD-1 inhibited collagen and ADP-induced platelet aggregation. This study proposes considering Serine Proteinase Associated Disintegrin (SPAD) as a new group of snake venom proteins. Members of this group contain a serine proteinase catalytic triad and a disintegrin-like motif. SPAD-1 caused visible morphological changes in MCF7 cells, including a reduction of the cell-to-cell attachments, rounding of cell shape and death, in vitro. SPAD-1 also showed a dose-dependent significant decrease in the invasive potency of breast cancer cells. Confocal microscopic analysis revealed the breakage of nuclei of the SPAD-1-treated cells. SPAD-1 also increased cell detachment from the poly L-lysine-coated, laminin-coated and fibronectin-coated culture plate matrices, confirming the disintegrin activity. This study concludes that SPAD-1 may be a good candidate for anti-tumour drug design in the future.


Subject(s)
Breast Neoplasms , Daboia , Animals , Humans , Female , Viper Venoms/chemistry , Disintegrins/pharmacology , Fibronectins , Serine Proteases/pharmacology , MCF-7 Cells , Laminin , Histidine , Aspartic Acid
6.
Hypertens Res ; 46(1): 50-62, 2023 01.
Article in English | MEDLINE | ID: mdl-36241707

ABSTRACT

Salt-sensitive hypertension is associated with poor clinical outcomes. The epithelial sodium channel (ENaC) in the kidney plays pivotal roles in sodium reabsorption and blood pressure regulation, in which its γ subunit is activated by extracellular serine proteases. In proteinuric nephropathies, plasmin filtered through injured glomeruli reportedly activates γENaC in the distal nephron and causes podocyte injury. We previously reported that Dahl salt-sensitive (DS) rats fed a high-salt (HS) diet developed hypertension and proteinuria along with γENaC activation and that a synthetic serine protease inhibitor, camostat mesilate, mitigated these changes. However, the role of plasmin in DS rats remained unclear. In this study, we evaluated the relationship between plasmin and hypertension as well as podocyte injury and the effects of plasmin inhibitors in DS rats. Five-week-old DS rats were divided into normal-salt diet, HS diet, and HS+plasmin inhibitor (either tranexamic acid [TA] or synthetic plasmin inhibitor YO-2) groups. After blood pressure measurement and 24 h urine collection over 5 weeks, rats were sacrificed for biochemical analyses. The HS group displayed severe hypertension and proteinuria together with activation of plasmin in urine and γENaC in the kidney, which was significantly attenuated by YO-2 but not TA. YO-2 inhibited the attachment of plasmin(ogen) to podocytes and alleviated podocyte injury by inhibiting apoptosis and inflammatory/profibrotic cytokines. YO-2 also suppressed upregulation of protease-activated receptor-1 and phosphorylated ERK1/2. These results indicate an important role of plasmin in the development of salt-sensitive hypertension and related podocyte injury, suggesting plasmin inhibition as a potential therapeutic strategy.


Subject(s)
Antifibrinolytic Agents , Hypertension , Podocytes , Rats , Animals , Rats, Inbred Dahl , Epithelial Sodium Channels , Fibrinolysin/pharmacology , Fibrinolysin/therapeutic use , Serine Proteases/pharmacology , Serine Proteases/therapeutic use , Antifibrinolytic Agents/pharmacology , Antifibrinolytic Agents/therapeutic use , Blood Pressure , Serine Endopeptidases , Sodium Chloride, Dietary/pharmacology , Proteinuria/complications
7.
An Acad Bras Cienc ; 94(3): e20211090, 2022.
Article in English | MEDLINE | ID: mdl-36259824

ABSTRACT

The search for new therapeutic strategies for leishmaniasis treatment is essential due to the side effects of available drugs and the increasing incidence of resistance to them. Marine sponges use chemical compounds as a defense mechanism, and several of them present interesting pharmacological properties. The aim of this study was to evaluate the in vitro activity of the aqueous extract of the marine sponge Dercitus (Stoeba) latex against Leishmania amazonensis. MIC and toxicity against mammal cells were evaluated through broth microdilution assays. Transmission electron microscopy analysis was performed to assess possible effects on L. amazonensis ultrastructure. Arginase and proteolytic activities were measured by spectrometric methodologies. The extract of Dercitus (Stoeba) latex displayed antileishmanial activity and moderate toxicity against peritonial macrophages. Ultrastructural changes were observed after the growth of L. amazonensis promastigotes in the presence of the extract at 150 µg.ml-1 (IC50), mainly on acidocalcysomes. The extract was able to inhibit the activity of arginase and serine proteases. This study shows that Dercitus (Stoeba) latex aqueous extract may be a novel potential source of protozoa protease inhibitors and drugs that are less toxic to be used in the treatment of L. amazonensis infections.


Subject(s)
Antiprotozoal Agents , Leishmania mexicana , Porifera , Animals , Latex/pharmacology , Arginase/pharmacology , Brazil , Leishmania mexicana/ultrastructure , Antiprotozoal Agents/pharmacology , Protease Inhibitors/pharmacology , Serine Proteases/pharmacology , Mammals
8.
Mol Oncol ; 16(19): 3568-3584, 2022 10.
Article in English | MEDLINE | ID: mdl-36056637

ABSTRACT

Previously, we found that telaprevir (Tel), the inhibitor of hepatitis C virus NS3/4A serine protease, reduces estrogen receptor α (ERα) content at the transcriptional level without binding to the receptor, prevents ERα transcriptional activity, and inhibits basal and 17ß-estradiol (E2)-dependent cell proliferation in different breast cancer (BC) cell lines. Here, we further characterize the Tel action mechanisms on ERα levels and function, identify a possible molecular target of Tel in BC cells, and evaluate Tel as an antiproliferative agent for BC treatment. Tel-dependent reduction in ERα levels and function depends on a Tel-dependent decrease in FOXA1 levels and activity. The effect of Tel is transduced by the IGF1-R/AKT/FOXA1 pathway, with the antiviral compound interacting with IGF1-R. Tel prevents the proliferation of several BC cell lines, while it does not affect the proliferation of normal nontransformed cell lines, and its antiproliferative effect is correlated with the ratio of FOXA1/IGF1-R expression. In conclusion, Tel interferes with the IGF1-R/AKT/FOXA1 pathway and induces cell death in ERα-expressing BC cells. Thus, we propose that this antiviral could be repurposed for the treatment of ERα-expressing BC.


Subject(s)
Breast Neoplasms , Estrogen Receptor alpha , Antiviral Agents/pharmacology , Breast Neoplasms/genetics , Cell Death , Cell Line, Tumor , Cell Proliferation , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , Oligopeptides , Proto-Oncogene Proteins c-akt/metabolism , Serine Proteases/metabolism , Serine Proteases/pharmacology , Serine Proteases/therapeutic use
9.
Neurobiol Aging ; 118: 106-107, 2022 10.
Article in English | MEDLINE | ID: mdl-35914472

ABSTRACT

One potential therapeutic strategy for Alzheimer disease (AD) is to promote degradation of amyloid beta (Aß) and we previously demonstrated that the lysosomal protease tripeptidyl peptidase 1 (TPP1) can degrade Aß fibrils in vitro. In this study, we tested the hypothesis that increasing levels of TPP1 might promote degradation of Aß under physiological conditions, slowing or preventing its accumulation in the brain with subsequent therapeutic benefits. We used 2 approaches to increase TPP1 activity in the brain of J20 mice, an AD model that accumulates Aß and exhibits cognitive defects: transgenic overexpression of TPP1 in the brain and a pharmacological approach employing administration of recombinant TPP1. While we clearly observed the expected AD phenotype of the J20 mice based on pathology and measurement of behavioral and cognitive defects, we found that elevation of TPP1 activity by either experimental approach failed to have any measurable beneficial effect on disease phenotype.


Subject(s)
Alzheimer Disease , Tripeptidyl-Peptidase 1 , Alzheimer Disease/pathology , Aminopeptidases/genetics , Aminopeptidases/metabolism , Aminopeptidases/pharmacology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Disease Models, Animal , Mice , Mice, Transgenic , Serine Proteases/genetics , Serine Proteases/metabolism , Serine Proteases/pharmacology
10.
Med Res Rev ; 42(6): 2126-2167, 2022 11.
Article in English | MEDLINE | ID: mdl-35762498

ABSTRACT

The rising pandemic caused by a coronavirus, resulted in a scientific quest to discover some effective treatments against its etiologic agent, the severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2). This research represented a significant scientific landmark and resulted in many medical advances. However, efforts to understand the viral mechanism of action and how the human body machinery is subverted during the infection are still ongoing. Herein, we contributed to this field with this compilation of the roles of both viral and human enzymes in the context of SARS-CoV-2 infection. In this sense, this overview reports that proteases are vital for the infection to take place: from SARS-CoV-2 perspective, the main protease (Mpro ) and papain-like protease (PLpro ) are highlighted; from the human body, angiotensin-converting enzyme-2, transmembrane serine protease-2, and cathepsins (CatB/L) are pointed out. In addition, the influence of the virus on other enzymes is reported as the JAK/STAT pathway and the levels of lipase, enzymes from the cholesterol metabolism pathway, amylase, aspartate aminotransferase, alanine aminotransferase, lactate dehydrogenase, and glyceraldehyde 3-phosphate dehydrogenase are also be disturbed in SARS-CoV-2 infection. Finally, this paper discusses the importance of detailed enzymatic studies for future treatments against SARS-CoV-2, and how some issues related to the syndrome treatment can create opportunities in the biotechnological market of enzymes and the development of new drugs.


Subject(s)
COVID-19 Drug Treatment , Alanine Transaminase/pharmacology , Amylases/pharmacology , Angiotensins/pharmacology , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Aspartate Aminotransferases/pharmacology , Cathepsins/pharmacology , Cholesterol , Human Body , Humans , Janus Kinases/pharmacology , Lactate Dehydrogenases , Lipase/pharmacology , Papain/pharmacology , SARS-CoV-2 , STAT Transcription Factors/pharmacology , Serine Proteases/pharmacology , Signal Transduction
11.
Nutr Cancer ; 74(1): 333-345, 2022.
Article in English | MEDLINE | ID: mdl-33544002

ABSTRACT

AIM: Hepatocellular carcinoma (HCC) is one of the most common malignant tumors. TKP is a serine protease extracted from the fruit of Trichosanthes kirilowii. We investigated the impact of TKP on the proliferation of HCC cells and its underlying mechanisms. METHODS: Bel-7402 and HepG2 cell viability and colony formation capacity were evaluated using MTT and colony formation assays, respectively. Glucose uptake and lactate production were determined using glucose and lactate assay kits. The mRNA expressions of GLUT1, PDK, LDHA, PKM2, ß-catenin, c-Myc, and HnRNPA1 were assessed using real-time PCR analysis. Protein expression and the distribution of PKM2 were examined by western blot assay. RESULTS: TKP significantly inhibited Bel-7402 and HepG2 cell survival and colony formation capacity. The IC50 values of TKP against Bel-7402 and HepG2 cells were 31.37 ± 1.33 and 27.41 ± 0.81 µg/mL, respectively. TKP restrained aerobic glycolysis. TKP decreased the expression level, nuclear protein level and pyruvate kinase activity of PKM2, whereas overexpression PKM2 reversed the suppression of TKP on glycolysis. TKP inhibited the ß-catenin/c-Myc/HnRNPA1 pathway. LiCl treatment partly rescued the inhibitory effects of TKP on PKM2, aerobic glycolysis, and cell viability. CONCLUSION: TKP suppresses HCC cell proliferation via blocking PKM2-dependent glycolysis, which is regulated by inhibiting the ß-catenin/c-Myc/HnRNPA1 pathway.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Trichosanthes , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation , Glycolysis , Humans , Liver Neoplasms/pathology , Oligopeptides , Serine Proteases/pharmacology
12.
Toxins (Basel) ; 13(12)2021 12 16.
Article in English | MEDLINE | ID: mdl-34941743

ABSTRACT

Bitis arietans is a snake of medical importance found throughout sub-Saharan Africa and in savannas and pastures of Morocco and western Arabia. The effects of its venom are characterized by local and systemic alterations, such as inflammation and cardiovascular and hemostatic disturbances, which can lead to victims' death or permanent disability. To better characterize the inflammatory process induced by this snake's venom, the participation of eicosanoids and PAF (platelet- activating factor) in this response were demonstrated in a previous study. In addition, edema and early increased vascular permeability followed by an accumulation of polymorphonuclear (PMN) cells in the peritoneal cavity were accompanied by the production of the eicosanoids LTB4, LTC4, TXB2, and PGE2, and local and systemic production of IL-6 and MCP-1. In this context, the present study focused on the identification of inflammatory mediators produced by human macrophages derived from THP-1 cells in response to Bitis arietans venom (BaV), and Kn-Ba, a serine protease purified from this venom. Here, we show that Kn-Ba, and even the less intensive BaV, induced the production of the cytokine TNF and the chemokines RANTES and IL-8. Only Kn-Ba was able to induce the production of IL-6, MCP-1, and IP-10, whereas PGE2 was produced only in response to BaV. Finally, the release of IL-1ß in culture supernatants suggests the activation of the inflammasomes by the venom of Bitis arietans and by Kn-Ba, which will be investigated in more detail in future studies.


Subject(s)
Cytokines/metabolism , Inflammation/metabolism , Macrophages/drug effects , Serine Proteases/pharmacology , Viper Venoms/chemistry , Viperidae/physiology , Animals , Cytokines/genetics , Gene Expression Regulation/drug effects , Humans , Serine Proteases/chemistry , Serine Proteases/metabolism , THP-1 Cells
13.
J Virol ; 95(19): e0086121, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34160253

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the viral pathogen causing the coronavirus disease 2019 (COVID-19) global pandemic. No effective treatment for COVID-19 has been established yet. The serine protease transmembrane protease serine 2 (TMPRSS2) is essential for viral spread and pathogenicity by facilitating the entry of SARS-CoV-2 into host cells. The protease inhibitor camostat, an anticoagulant used in the clinic, has potential anti-inflammatory and antiviral activities against COVID-19. However, the potential mechanisms of viral resistance and antiviral activity of camostat are unclear. Herein, we demonstrate high inhibitory potencies of camostat for a panel of serine proteases, indicating that camostat is a broad-spectrum inhibitor of serine proteases. In addition, we determined the crystal structure of camostat in complex with a serine protease (uPA [urokinase-type plasminogen activator]), which reveals that camostat is inserted in the S1 pocket of uPA but is hydrolyzed by uPA, and the cleaved camostat covalently binds to Ser195. We also generated a homology model of the structure of the TMPRSS2 serine protease domain. The model shows that camostat uses the same inhibitory mechanism to inhibit the activity of TMPRSS2, subsequently preventing SARS-CoV-2 spread. IMPORTANCE Serine proteases are a large family of enzymes critical for multiple physiological processes and proven diagnostic and therapeutic targets in several clinical indications. The serine protease transmembrane protease serine 2 (TMPRSS2) was recently found to mediate SARS-CoV-2 entry into the host. Camostat mesylate (FOY 305), a serine protease inhibitor active against TMPRSS2 and used for the treatment of oral squamous cell carcinoma and chronic pancreatitis, inhibits SARS-CoV-2 infection of human lung cells. However, the direct inhibition mechanism of camostat mesylate for TMPRSS2 is unclear. Herein, we demonstrate that camostat uses the same inhibitory mechanism to inhibit the activity of TMPRSS2 as uPA, subsequently preventing SARS-CoV-2 spread.


Subject(s)
Antiviral Agents/pharmacology , Esters/pharmacology , Guanidines/pharmacology , SARS-CoV-2/drug effects , Serine Endopeptidases/chemistry , Serine Endopeptidases/pharmacology , Serine Proteases/pharmacology , Antiviral Agents/chemistry , COVID-19/prevention & control , Carcinoma, Squamous Cell , Esters/chemistry , Esters/metabolism , Guanidines/chemistry , Guanidines/metabolism , Humans , Molecular Dynamics Simulation , Mouth Neoplasms , Protein Domains , Sequence Alignment , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Serine Proteases/chemistry , Serine Proteases/metabolism , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/pharmacology , Virus Internalization/drug effects , COVID-19 Drug Treatment
14.
Biotechnol Lett ; 43(9): 1787-1798, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34028659

ABSTRACT

OBJECTIVE: Mycobacterial acid-resistant protease (MarP) is a membrane-associated serine protease involved in the survival of Mycobacterium tuberculosis in macrophages; here we produced MarP in the yeast Pichia pastoris and study its involvement in macrophage immune modulation. RESULTS: Pichia pastoris vectors, harboring a full-length or a partial sequence of MarP, were constructed. GS115 clones were selected, and homologous recombination at the AOX1 locus was assessed by PCR. Protein was purified by nickel affinity chromatography, and its effect on the cytokine profile was tested in human monocytes. Only the partial MarP protein (121-397 a.a.) lacking the transmembrane domain was successfully expressed as an N-glycosylated proteolytically active protease. In vitro stimulation of THP-1 cells with MarP promoted the release of TNF-α and IL-10. CONCLUSION: Mycobacterial MarP was successfully expressed in P. pastoris, and it is capable of cytokine release in vitro.


Subject(s)
Mycobacterium tuberculosis/enzymology , Pichia/growth & development , Serine Proteases/genetics , Serine Proteases/metabolism , Aldehyde Oxidase/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Chromatography, Affinity , Fungal Proteins/genetics , Gene Expression Regulation/drug effects , Homologous Recombination , Humans , Interleukin-10/metabolism , Monocytes/cytology , Monocytes/drug effects , Monocytes/immunology , Mycobacterium tuberculosis/genetics , Pichia/genetics , Pichia/metabolism , Protein Domains , Protein Engineering , Serine Proteases/chemistry , Serine Proteases/pharmacology , THP-1 Cells , Tumor Necrosis Factor-alpha/metabolism
15.
J Ethnopharmacol ; 276: 114170, 2021 Aug 10.
Article in English | MEDLINE | ID: mdl-33932515

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Sapindus saponaria, also popularly known as soapberry, has been used in folk medicinal values because of its therapeutic properties and several compounds in its composition, which represent a target in potential for drug discovery. However, few data about its potential toxicity has been reported. AIM OF THE STUDY: Plant proteins can perform essential roles in survival, acting as defense mechanism, as well functioning as important molecular reserves for its natural metabolism. The aim of the current study was to investigate the in vitro toxicity profile of protein extract of S. saponaria and detect protein potentially involved in biological effects such as collagen hydrolysis and inhibition of viral proteases. MATERIALS AND METHODS: Protein extract of soapberry seeds was investigated for its cytotoxic and genotoxic action using the Ames test. The protein extract was also subjected to a partial purification process of a protease and a protease inhibitor by gel chromatography filtration techniques and the partially isolated proteins were characterized biochemically. RESULTS: Seed proteins extract of S. saponaria was evaluated until 100 µg/mL concentration, presenting cytotoxicity and mutagenicity in bacterial model mostly when exposed to exogenous metabolic system and causing cytotoxic and genotoxic effects in HepG2 cells. The purification and partial characterization of a serine protease (43 kDa) and a cysteine protease inhibitor (32.8 kDa) from protein extract of S. Saponaria, corroborate the idea of ​​the biological use of the plant as an insecticide and larvicide. Although it shows cytotoxic, mutagenic and genotoxic effects. CONCLUSION: The overall results of the present study provide supportive data on the potential use of proteins produced in S. saponaria seeds as pharmacological and biotechnological agents that can be further explored for the development of new drugs.


Subject(s)
DNA Damage/drug effects , Plant Extracts/pharmacology , Plant Extracts/toxicity , Sapindus/chemistry , Seeds/chemistry , Biochemical Phenomena , Cell Death/drug effects , Cystatins/chemistry , Cystatins/isolation & purification , Cystatins/pharmacology , Hep G2 Cells , Humans , Lethal Dose 50 , Micronucleus Tests , Mutagenicity Tests , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Salmonella typhimurium/drug effects , Serine Proteases/chemistry , Serine Proteases/isolation & purification , Serine Proteases/pharmacology
16.
Biochim Biophys Acta Mol Cell Res ; 1868(3): 118925, 2021 03.
Article in English | MEDLINE | ID: mdl-33333088

ABSTRACT

Plant latex proteases (PLPs) are pharmacologically essential and are integral components of traditional medicine in the management of bleeding wounds. PLPs are known to promote blood coagulation and stop bleeding by interfering at various stages of hemostasis. There are a handful of scientific reports on thrombin-like enzymes characterized from plant latices. However, the role of plant latex thrombin-like enzymes in platelet aggregation is not well known. In the present study, we attempted to purify and characterize thrombin-like protease responsible for platelet aggregation. Among tested plant latices, Euphorbia genus latex protease fractions (LPFs) induced platelet aggregation. In Euphorbia genus, E. antiquorum LPF (EaLPF) strongly induced platelet aggregation and attenuated bleeding in mice. The purified thrombin-like serine protease, antiquorin (Aqn) is a glycoprotein with platelet aggregating activities that interfere in intrinsic and common pathways of blood coagulation cascade and alleviates bleeding and enhanced excision wound healing in mice. In continuation, the pharmacological inhibitor of PAR1 inhibited Aqn-induced phosphorylation of cPLA2, Akt, and P38 in human platelets. Moreover, Aqn-induced platelet aggregation was inhibited by pharmacological inhibitors of PAR1, PI3K, and P38. These data indicate that PAR1-Akt/P38 signaling pathways are involved in Aqn-induced platelet aggregation. The findings of the present study may open up a new avenue for exploiting Aqn in the treatment of bleeding wounds.


Subject(s)
Euphorbia/chemistry , Hemorrhage/drug therapy , Latex/chemistry , Serine Proteases/administration & dosage , Adult , Animals , Blood Coagulation , Disease Models, Animal , HEK293 Cells , Hemorrhage/etiology , Humans , Male , Mice , Phosphorylation , Plant Proteins/administration & dosage , Plant Proteins/pharmacology , Serine Proteases/pharmacology , Signal Transduction/drug effects , Wound Healing/drug effects , Young Adult
17.
Int J Biol Macromol ; 168: 631-639, 2021 Jan 31.
Article in English | MEDLINE | ID: mdl-33227332

ABSTRACT

A novel fibrinolytic enzyme, ACase was isolated from fruiting bodies of a mushroom, Agrocybe aegerita. ACase was purified by using ammonium sulfate precipitation, gel filtration, ion exchange and hydrophobic chromatographies to 237.12 fold with a specific activity of 1716.77 U/mg. ACase was found to be a heterodimer with molecular mass of 31.4 and 21.2 kDa by SDS-PAGE and appeared as a single band on Native-PAGE and fibrin-zymogram. The N-terminal sequence of the two subunits of ACase was AIVTQTNAPWGL (subunit 1) and SNADGNGHGTHV (subunit 2). ACase had maximal activity at 47 °C and pH 7.6. It's activity was improved by Cu2+, Na+, Fe3+, Zn2+, Ba2+, K+ and Mn2+, but inhibited by Fe2+, Mg2+ and Ca2+. PMSF, SBTI, aprotinine and Lys inhibited the enzyme activity, which suggested that ACase was a serine protease. ACase could degrade all three chains (α, ß and γ) of fibrinogen. Moreover, the enzyme acted as both, a plasmin-like fibrinolytic enzyme and a plasminogen activator. It could hydrolyze human thrombin slightly, which indicated that the ACase could inhibit the activity of thrombin and acted as an anticoagulant to prevent thrombosis. Based on these results, ACase might act as a therapeutic agent for treating thrombosis, or as a functional food. Further investigation of the enzyme is underway.


Subject(s)
Agrocybe/enzymology , Anticoagulants/pharmacology , Fibrinolytic Agents/pharmacology , Serine Proteases/pharmacology , Amino Acid Sequence , Anticoagulants/chemistry , Anticoagulants/isolation & purification , Chemical Phenomena , Chromatography, Ion Exchange , Fibrinogen/metabolism , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/isolation & purification , Fruiting Bodies, Fungal/enzymology , Fungal Proteins/chemistry , Fungal Proteins/isolation & purification , Fungal Proteins/pharmacology , Humans , Hydrogen-Ion Concentration , Immunoglobulin G/metabolism , Molecular Weight , Protein Multimerization , Serine Proteases/chemistry , Serine Proteases/isolation & purification , Serum Albumin, Human/metabolism , Thrombin/metabolism
18.
Molecules ; 25(15)2020 Jul 28.
Article in English | MEDLINE | ID: mdl-32731325

ABSTRACT

Chronic wounds are a major health problem that cause millions of dollars in expenses every year. Among all the treatments used, active wound treatments such as enzymatic treatments represent a cheaper and specific option with a fast growth category in the market. In particular, bacterial and plant proteases have been employed due to their homology to human proteases, which drive the normal wound healing process. However, the use of these proteases has demonstrated results with low reproducibility. Therefore, alternative sources of proteases such as snake venom have been proposed. Here, we performed a functional mining of proteases from rattlesnakes (Crotalus ornatus, C. molossus nigrescens, C. scutulatus, and C. atrox) due to their high protease predominance and similarity to native proteases. To characterize Crotalus spp. Proteases, we performed different protease assays to measure and confirm the presence of metalloproteases and serine proteases, such as the universal protease assay and zymography, using several substrates such as gelatin, casein, hemoglobin, L-TAME, fibrinogen, and fibrin. We found that all our venom extracts degraded casein, gelatin, L-TAME, fibrinogen, and fibrin, but not hemoglobin. Crotalus ornatus and C. m. nigrescens extracts were the most proteolytic venoms among the samples. Particularly, C. ornatus predominantly possessed low molecular weight proteases (P-I metalloproteases). Our results demonstrated the presence of metalloproteases capable of degrading gelatin (a collagen derivative) and fibrin clots, whereas serine proteases were capable of degrading fibrinogen-generating fibrin clots, mimicking thrombin activity. Moreover, we demonstrated that Crotalus spp. are a valuable source of proteases that can aid chronic wound-healing treatments.


Subject(s)
Crotalid Venoms/enzymology , Crotalus/metabolism , Metalloproteases , Reptilian Proteins , Serine Proteases , Wounds and Injuries/drug therapy , Animals , Fibrinolysis/drug effects , Humans , Metalloproteases/chemistry , Metalloproteases/pharmacology , Reproducibility of Results , Reptilian Proteins/chemistry , Reptilian Proteins/pharmacology , Serine Proteases/chemistry , Serine Proteases/pharmacology , Wounds and Injuries/metabolism , Wounds and Injuries/pathology
19.
Nucleic Acid Ther ; 30(5): 276-288, 2020 10.
Article in English | MEDLINE | ID: mdl-32486960

ABSTRACT

Activated protein C (APC) is a serine protease with anticoagulant and cytoprotective activities. Nonanticoagulant APC mutants show beneficial effects as cytoprotective agents. To study, if such biased APC signaling can be achieved by APC-binding ligands, the aptamer technology has been used. A G-quadruplex-containing aptamer, G-NB3, has been selected that binds to the basic exosite of APC with a KD of 0.2 nM and shows no binding to APC-related serine proteases or the zymogen protein C. G-NB3 inhibits the inactivation of activated cofactors V and VIII with IC50 values of 11.6 and 13.1 nM, respectively, without inhibiting the cytoprotective and anti-inflammatory functions of APC as tested using a staurosporine-induced apoptosis assay and a vascular barrier protection assay. In addition, G-NB3 prolongs the plasma half-life of APC through inhibition of APC-serine protease inhibitor complex formation. These physicochemical and functional characteristics qualify G-NB3 as a promising therapeutic agent usable to enhance the cytoprotective functions of APC without increasing the risk of APC-related hemorrhage.


Subject(s)
Aptamers, Nucleotide/pharmacology , Hemorrhage/drug therapy , Protein C/pharmacology , Serine Proteases/pharmacology , Anticoagulants/pharmacology , Aptamers, Nucleotide/genetics , Blood Coagulation/drug effects , G-Quadruplexes , Hemorrhage/pathology , Humans , Ligands , Protein Binding/genetics , Protein C/genetics , Serine Proteases/genetics , Signal Transduction/drug effects , Thrombin/genetics
20.
Int Immunopharmacol ; 86: 106699, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32570037

ABSTRACT

Inflammatory bowel disease (IBD), which includes ulcerative colitis (UC) and Crohn's disease (CD), is a chronic autoimmune disease. At present, worms and their products has been shown to have protective effects on immune-mediated diseases. Therefore, we aimed to investigate the effect of the recombination Trichinella spiralis (T. spiralis, Ts) adult serine protease-like protein rTs-ADSp-7 on a 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced CD mouse model. Colitis was induced by intrarectal administration of a TNBS solution. The disease activity index (DAI), which included weight loss, diarrhoea, and bloody stool, was measured. Colon segments were stained with haematoxylin and eosin (H.E.) for histopathological score. Cytokine release in the serum was analysed by meso scale discovery (MSD). Cytokine release in the colon was detected by ELISA. Splenocytes were separated, and the cytokine profiles of Th1 (IFN-γ), Th2 (IL-4), Th17 (IL-17A) and Treg cells were analysed by flow cytometry. Our result showed that rTs-ADSp-7 reduced the clinical disease activity of TNBS-induced colitis in mice. In addition, we found that rTs-ADSp-7 reduced the production of Th1- and Th17-related cytokines while upregulating the expression of Th2- and Treg-related cytokines in TNBS-induced colitis mice. rTs-ADSp-7 also increased the population of Th2 and Treg cells in TNBS-induced colitis mice. rTs-ADSp-7 alleviated the severity of TNBS-induced colitis while balancing the CD4+ T cell immune response. rTs-ADSp-7 has therapeutic potential for colitis treatment and can be used as a helminth-derived protein therapy for CD or other Th1 immunity-mediated diseases.


Subject(s)
Colitis/drug therapy , Colitis/immunology , Helminth Proteins/pharmacology , Serine Proteases/pharmacology , Trichinella spiralis/enzymology , Aging , Animals , Colitis/chemically induced , Colon/immunology , Colon/pathology , Crohn Disease/chemically induced , Crohn Disease/drug therapy , Crohn Disease/immunology , Cytokines/blood , Cytokines/metabolism , Disease Models, Animal , Female , Helminth Proteins/therapeutic use , Mice, Inbred BALB C , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Serine Proteases/therapeutic use , Spleen/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Th2 Cells/immunology , Trinitrobenzenesulfonic Acid/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...