guanidine alkaloids
Recently Published Documents


TOTAL DOCUMENTS

73
(FIVE YEARS 8)

H-INDEX

22
(FIVE YEARS 2)

2021 ◽  
Vol 12 ◽  
Author(s):  
Juan A. Rubiolo ◽  
Emilio Lence ◽  
Concepción González-Bello ◽  
María Roel ◽  
José Gil-Longo ◽  
...  

Crambescins are guanidine alkaloids from the sponge Crambe crambe. Crambescin C1 (CC) induces metallothionein genes and nitric oxide (NO) is one of the triggers. We studied and compared the in vitro, in vivo, and in silico effects of some crambescine A and C analogs. HepG2 gene expression was analyzed using microarrays. Vasodilation was studied in rat aortic rings. In vivo hypotensive effect was directly measured in anesthetized rats. The targets of crambescines were studied in silico. CC and homo-crambescine C1 (HCC), but not crambescine A1 (CA), induced metallothioneins transcripts. CC increased NO production in HepG2 cells. In isolated rat aortic rings, CC and HCC induced an endothelium-dependent relaxation related to eNOS activation and an endothelium-independent relaxation related to iNOS activation, hence both compounds increase NO and reduce vascular tone. In silico analysis also points to eNOS and iNOS as targets of Crambescin C1 and source of NO increment. CC effect is mediated through crambescin binding to the active site of eNOS and iNOS. CC docking studies in iNOS and eNOS active site revealed hydrogen bonding of the hydroxylated chain with residues Glu377 and Glu361, involved in the substrate recognition, and explains its higher binding affinity than CA. The later interaction and the extra polar contacts with its pyrimidine moiety, absent in the endogenous substrate, explain its role as exogenous substrate of NOSs and NO production. Our results suggest that CC serve as a basis to develop new useful drugs when bioavailability of NO is perturbed.


Biomolecules ◽  
2021 ◽  
Vol 11 (3) ◽  
pp. 460
Author(s):  
Amr El-Demerdash ◽  
Ahmed M. Metwaly ◽  
Afnan Hassan ◽  
Tarek Mohamed Abd El-Aziz ◽  
Eslam B. Elkaeed ◽  
...  

The huge global expansion of the COVID-19 pandemic caused by the novel SARS-corona virus-2 is an extraordinary public health emergency. The unavailability of specific treatment against SARS-CoV-2 infection necessitates the focus of all scientists in this direction. The reported antiviral activities of guanidine alkaloids encouraged us to run a comprehensive in silico binding affinity of fifteen guanidine alkaloids against five different proteins of SARS-CoV-2, which we investigated. The investigated proteins are COVID-19 main protease (Mpro) (PDB ID: 6lu7), spike glycoprotein (PDB ID: 6VYB), nucleocapsid phosphoprotein (PDB ID: 6VYO), membrane glycoprotein (PDB ID: 6M17), and a non-structural protein (nsp10) (PDB ID: 6W4H). The binding energies for all tested compounds indicated promising binding affinities. A noticeable superiority for the pentacyclic alkaloids particularly, crambescidin 786 (5) and crambescidin 826 (13) has been observed. Compound 5 exhibited very good binding affinities against Mpro (ΔG = −8.05 kcal/mol), nucleocapsid phosphoprotein (ΔG = −6.49 kcal/mol), and nsp10 (ΔG = −9.06 kcal/mol). Compound 13 showed promising binding affinities against Mpro (ΔG = −7.99 kcal/mol), spike glycoproteins (ΔG = −6.95 kcal/mol), and nucleocapsid phosphoprotein (ΔG = −8.01 kcal/mol). Such promising activities might be attributed to the long ω-fatty acid chain, which may play a vital role in binding within the active sites. The correlation of c Log P with free binding energies has been calculated. Furthermore, the SAR of the active compounds has been clarified. The Absorption, Distribution, Metabolism, Excretion, and Toxicity (ADMET) studies were carried out in silico for the 15 compounds; most examined compounds showed optimal to good range levels of ADMET aqueous solubility, intestinal absorption and being unable to pass blood brain barrier (BBB), non-inhibitors of CYP2D6, non-hepatotoxic, and bind plasma protein with a percentage less than 90%. The toxicity of the tested compounds was screened in silico against five models (FDA rodent carcinogenicity, carcinogenic potency TD50, rat maximum tolerated dose, rat oral LD50, and rat chronic lowest observed adverse effect level (LOAEL)). All compounds showed expected low toxicity against the tested models. Molecular dynamic (MD) simulations were also carried out to confirm the stable binding interactions of the most promising compounds, 5 and 13, with their targets. In conclusion, the examined 15 alkaloids specially 5 and 13 showed promising docking, ADMET, toxicity and MD results which open the door for further investigations for them against SARS-CoV-2.


2020 ◽  
Author(s):  
Amr El-Demerdash ◽  
Ahmed M. Metwaly ◽  
Tarek Mohamed Abd El-Aziz4 ◽  
Ibrahim H. Eissa ◽  
James D. Stockand

<p>A comprehensive <i>in silico</i> binding affinity of fifteen guanidine alkaloids against five different proteins of SARS-CoV-2 has been investigated. The investigated proteins are COVID-19 main protease (M<sup>pro</sup>) (PDB ID: 6lu7), spike glycoprotein (PDB ID: 6VYB), nucleocapsid phosphoprotein (PDB ID: 6VYO), membrane glycoprotein (PDB ID: 6M17), and non-structural protein (nsp10) (PDB ID: 6W4H). The binding energies for all tested compounds indicated promising binding affinities. A noticeable superiority for the pentacyclic alkaloids particularly, crambescidin 786 (<b>5</b>)<b> </b>and<b> </b>crambescidin 826<b> </b>(<b>13</b>) have been observed. Compound <b>5</b> exhibited very good binding affinities against M<sup>pro</sup> (ΔG = -8.05 kcal/mol), nucleocapsid phosphoprotein (ΔG = -6.49 kcal/mol), and nsp10 (ΔG = -9.06 kcal/mol). Compound <b>13</b> showed promising binding affinities against M<sup>pro</sup> (ΔG = -7.99 kcal/mol), spike glycoproteins (ΔG = -6.95 kcal/mol), and nucleocapsid phosphoprotein (ΔG = -8.01 kcal/mol). Such promising activities might be attributed to the long ω-fatty acid chain, which may play a vital role in binding within the active sites. The ADMET studies were carried out <i>in silico</i> for the 15 compounds, all examined compounds (except compounds <b>8</b> and <b>15</b>) have low or very low BBB penetration levels. Compounds <b>1</b>, <b>5</b>, <b>6</b>, <b>9</b>, <b>12</b> and <b>13</b> showed optimal range levels of ADMET aqueous solubility. Compounds <b>1</b>, <b>2</b>, <b>3</b>, <b>8</b>, and <b>15</b> were predicted to have good intestinal absorption levels, while compounds <b>4</b>, <b>7</b>, <b>9</b>, <b>10</b>, and <b>14</b> showed moderate absorption levels. All examined alkaloids (except the bicyclic compound <b>8</b>) were predicted not to be inhibitors of CYP2D6, non-hepatotoxic, and bind plasma protein with a percentage less than 90%. The toxicity of the tested compounds was screened <i>in silico</i> against five models (FDA rodent carcinogenicity, carcinogenic potency TD<sub>50</sub>, rat maximum tolerated dose, rat oral LD<sub>50</sub> and rat chronic LOAEL). All compounds showed expected low toxicity against the tested models. </p>


2020 ◽  
Author(s):  
Amr El-Demerdash ◽  
Ahmed M. Metwaly ◽  
Tarek Mohamed Abd El-Aziz4 ◽  
Ibrahim H. Eissa ◽  
James D. Stockand

<p>A comprehensive <i>in silico</i> binding affinity of fifteen guanidine alkaloids against five different proteins of SARS-CoV-2 has been investigated. The investigated proteins are COVID-19 main protease (M<sup>pro</sup>) (PDB ID: 6lu7), spike glycoprotein (PDB ID: 6VYB), nucleocapsid phosphoprotein (PDB ID: 6VYO), membrane glycoprotein (PDB ID: 6M17), and non-structural protein (nsp10) (PDB ID: 6W4H). The binding energies for all tested compounds indicated promising binding affinities. A noticeable superiority for the pentacyclic alkaloids particularly, crambescidin 786 (<b>5</b>)<b> </b>and<b> </b>crambescidin 826<b> </b>(<b>13</b>) have been observed. Compound <b>5</b> exhibited very good binding affinities against M<sup>pro</sup> (ΔG = -8.05 kcal/mol), nucleocapsid phosphoprotein (ΔG = -6.49 kcal/mol), and nsp10 (ΔG = -9.06 kcal/mol). Compound <b>13</b> showed promising binding affinities against M<sup>pro</sup> (ΔG = -7.99 kcal/mol), spike glycoproteins (ΔG = -6.95 kcal/mol), and nucleocapsid phosphoprotein (ΔG = -8.01 kcal/mol). Such promising activities might be attributed to the long ω-fatty acid chain, which may play a vital role in binding within the active sites. The ADMET studies were carried out <i>in silico</i> for the 15 compounds, all examined compounds (except compounds <b>8</b> and <b>15</b>) have low or very low BBB penetration levels. Compounds <b>1</b>, <b>5</b>, <b>6</b>, <b>9</b>, <b>12</b> and <b>13</b> showed optimal range levels of ADMET aqueous solubility. Compounds <b>1</b>, <b>2</b>, <b>3</b>, <b>8</b>, and <b>15</b> were predicted to have good intestinal absorption levels, while compounds <b>4</b>, <b>7</b>, <b>9</b>, <b>10</b>, and <b>14</b> showed moderate absorption levels. All examined alkaloids (except the bicyclic compound <b>8</b>) were predicted not to be inhibitors of CYP2D6, non-hepatotoxic, and bind plasma protein with a percentage less than 90%. The toxicity of the tested compounds was screened <i>in silico</i> against five models (FDA rodent carcinogenicity, carcinogenic potency TD<sub>50</sub>, rat maximum tolerated dose, rat oral LD<sub>50</sub> and rat chronic LOAEL). All compounds showed expected low toxicity against the tested models. </p>


2020 ◽  
pp. 1-5
Author(s):  
Caroline Killian ◽  
Shelby L. Johnson ◽  
Hang Ma ◽  
Brendan McKeown ◽  
Luke McDougall ◽  
...  
Keyword(s):  

Marine Drugs ◽  
2019 ◽  
Vol 17 (11) ◽  
pp. 598 ◽  
Author(s):  
H. Herath ◽  
Sarah Preston ◽  
Abdul Jabbar ◽  
Jose Garcia-Bustos ◽  
Aya Taki ◽  
...  

There is an urgent need to discover and develop new anthelmintics for the treatment of parasitic nematodes of veterinary importance to circumvent challenges linked to drug resistant parasites. Being one of the most diverse natural ecosystems, the marine environment represents a rich resource of novel chemical entities. This study investigated 2000 extracts from marine invertebrates, collected from Australian waters, for anthelmintic activity. Using a well-established in vitro bioassay, these extracts were screened for nematocidal activity against Haemonchus contortus — a socioeconomically important parasitic nematode of livestock animals. Extracts (designated Mu-1, Ha-1 and Ha-2) from two marine sponges (Monanchora unguiculata and Haliclona sp.) each significantly affected larvae of H. contortus. Individual extracts displayed a dose-dependent inhibition of both the motility of exsheathed third-stage larvae (xL3s) and the development of xL3s to fourth-stage larvae (L4s). Active fractions in each of the three extracts were identified using bioassay-guided fractionation. From the active fractions from Monanchora unguiculata, a known pentacyclic guanidine alkaloid, fromiamycalin (1), was purified. This alkaloid was shown to be a moderately potent inhibitor of L4 development (half-maximum inhibitory concentration (IC50) = 26.6 ± 0.74 µM) and L4 motility (IC50 = 39.4 ± 4.83 µM), although it had a relatively low potency at inhibiting of xL3 motility (IC50 ≥ 100 µM). Investigation of the active fractions from the two Haliclona collections led to identification of a mixture of amino alcohol lipids, and, subsequently, a known natural product halaminol A (5). Anthelmintic profiling showed that 5 had limited potency at inhibiting larval development and motility. These data indicate that fromiamycalin, other related pentacyclic guanidine alkaloids and/or halaminols could have potential as anthelmintics following future medicinal chemistry efforts.


Marine Drugs ◽  
2019 ◽  
Vol 17 (1) ◽  
pp. 22 ◽  
Author(s):  
Irina Bakunina ◽  
Galina Likhatskaya ◽  
Lubov Slepchenko ◽  
Larissa Balabanova ◽  
Liudmila Tekutyeva ◽  
...  

The effect of monanchomycalin B, monanhocicidin A, and normonanhocidin A isolated from the Northwest Pacific sample of the sponge Monanchora pulchra was investigated on the activity of α-galactosidase from the marine γ-proteobacterium Pseudoalteromonas sp. KMM 701 (α-PsGal), and α-N-acetylgalactosaminidase from the marine bacterium Arenibacter latericius KMM 426T (α-NaGa). All compounds are slow-binding irreversible inhibitors of α-PsGal, but have no effect on α-NaGa. A competitive inhibitor d-galactose protects α-PsGal against the inactivation. The inactivation rate (kinact) and equilibrium inhibition (Ki) constants of monanchomycalin B, monanchocidin A, and normonanchocidin A were 0.166 ± 0.029 min−1 and 7.70 ± 0.62 μM, 0.08 ± 0.003 min−1 and 15.08 ± 1.60 μM, 0.026 ± 0.000 min−1, and 4.15 ± 0.01 μM, respectively. The 2D-diagrams of α-PsGal complexes with the guanidine alkaloids were constructed with “vessel” and “anchor” parts of the compounds. Two alkaloid binding sites on the molecule of α-PsGal are shown. Carboxyl groups of the catalytic residues Asp451 and Asp516 of the α-PsGal active site interact with amino groups of “anchor” parts of the guanidine alkaloid molecules.


2018 ◽  
Vol 58 (2) ◽  
pp. 520-525 ◽  
Author(s):  
Siguara B. L. Silva ◽  
François Oberhänsli ◽  
Marie-Aude Tribalat ◽  
Grégory Genta-Jouve ◽  
Jean-Louis Teyssié ◽  
...  

2018 ◽  
Vol 131 (2) ◽  
pp. 530-535
Author(s):  
Siguara B. L. Silva ◽  
François Oberhänsli ◽  
Marie-Aude Tribalat ◽  
Grégory Genta-Jouve ◽  
Jean-Louis Teyssié ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document