Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 267
Filtrar
1.
J Zhejiang Univ Sci B ; 25(9): 773-788, 2024 Sep 15.
Artigo em Inglês, Chinês | MEDLINE | ID: mdl-39308067

RESUMO

The use of nitrification inhibitors has been suggested as a strategy to decrease cadmium (Cd) accumulation in crops. However, the most efficient nitrification inhibitor for mitigating crop Cd accumulation remains to be elucidated, and whether and how changes in soil microbial structure are involved in this process also remains unclear. To address these questions, this study applied three commercial nitrification inhibitors, namely, dicyandiamide (DCD), 3,4-dimethylpyrazole phosphate (DMPP), and nitrapyrin (NP), to pakchoi. The results showed that both DCD and DMPP (but not NP) could efficiently decrease Cd concentrations in pakchoi in urea- and ammonium-fertilized soils. In addition, among the three tested nitrification inhibitors, DMPP was the most efficient in decreasing the Cd concentration in pakchoi. The nitrification inhibitors decreased pakchoi Cd concentrations by suppressing acidification-induced Cd availability and reshaping the soil microbial structure; the most effective nitrification inhibitor was DMPP. Ammonia oxidation generates the most protons during nitrification and is inhibited by nitrification inhibitors. Changes in environmental factors and predatory bacterial abundance caused by the nitrification inhibitors changed the soil microbial structure and increased the potential participants in plant Cd accumulation. In summary, our study identified DMPP as the most efficient nitrification inhibitor for mitigating crop Cd contamination and observed that the soil microbial structural changes caused by the nitrification inhibitors contributed to decreasing Cd concentration in pakchoi.


Assuntos
Cádmio , Guanidinas , Nitrificação , Microbiologia do Solo , Cádmio/metabolismo , Nitrificação/efeitos dos fármacos , Guanidinas/metabolismo , Guanidinas/farmacologia , Poluentes do Solo/metabolismo , Pirazóis/farmacologia , Solo/química , Fertilizantes , Amônia/metabolismo
2.
Biochem Biophys Res Commun ; 733: 150603, 2024 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-39216203

RESUMO

Serine proteases are among the important groups of enzymes having significant roles in cell biology. Trypsin is a representative member of the serine superfamily of enzymes, produced by acinar cells of pancreas. It is a validated drug target for various ailments including pancreatitis and colorectal cancer. Premature activation of trypsin is involved in the lysis of pancreatic tissues, which causes pancreatitis. It is also reported to be involved in colorectal carcinoma by activating other proteases, such as matrix metalloproteinase (MMPs). The development of novel trypsin inhibitors with good pharmacokinetic properties could play important roles in pharmaceutical sciences. This study reports the crystal structures of bovine pancreatic trypsin with four molecules; cimetidine, famotidine, pimagedine, and guanidine. These compounds possess binding affinity towards the active site (S1) of trypsin. The structures of all four complexes provided insight of the binding of four different ligands, as well as the dynamics of the active site towards the bind with different size ligands. This study might be helpful in designing of new potent inhibitors of trypsin and trypsin like serine proteases.


Assuntos
Cimetidina , Famotidina , Tripsina , Tripsina/metabolismo , Tripsina/química , Famotidina/química , Famotidina/metabolismo , Animais , Cimetidina/metabolismo , Cimetidina/química , Cimetidina/farmacologia , Bovinos , Ligação Proteica , Guanidina/química , Guanidina/metabolismo , Cristalografia por Raios X , Modelos Moleculares , Domínio Catalítico , Serina Proteases/metabolismo , Serina Proteases/química , Inibidores da Tripsina/metabolismo , Inibidores da Tripsina/química , Sítios de Ligação , Conformação Proteica , Guanidinas/metabolismo , Guanidinas/química
3.
Sci Total Environ ; 941: 173257, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-38761944

RESUMO

The combined exposure of heavy metals and organic contaminates can influence the transport and accumulation of heavy metals within the soil-rice system. However, the underlying mechanisms of this process remain largely unknown. Herein, this study investigated the influence of three neonicotinoid insecticides (NIs), including imidacloprid (IMI), clothianidin (CLO), and thiamethoxam (THI), on the Cd transport and accumulation in rice (Oryza sativa) at different growth stages. Particular focus lied on their complex interaction and key genes expression involved in Cd transport. Results showed that the interaction between Cd and NIs was the dominant factor affecting Cd transport and accumulation in rice exposed to NIs. All three NIs chelated with Cd with nitrogen (N) on the IMI and THI nitro groups, and the N on the CLO nitro guanidine group. Interestingly, this chelation behavior varied between the tillering stage and the filling/ripening stages, resulting in diverse patterns of Cd accumulation in rice tissues. During the tillering stage, all three NIs considerably inhibited Cd bioavailability and transport to the above-ground part, lowering Cd content in the stem and leaf. The inhibition was increased with stronger chelation ability in the order of IMI (-0.46 eV) > CLO (-0.41 eV) > THI (-0.11 eV), with IMI exhibiting the highest binding energy for Cd and reducing Cd transfers from root to stem by an impressive 94.49 % during the tillering stage. Conversely, during the filling/ripening stages, NIs facilitated Cd accumulation in rice roots, stems, leaves, and grains. This was mainly attributed to the generation of nitrate ions and the release of Cd2+ during the chelation between Cd and NIs under drainage condition. These findings provide theoretical basis for the treatment of combined contamination in field and deep insights into understanding the interaction of organic contaminants with heavy metals in rice culture process.


Assuntos
Cádmio , Inseticidas , Neonicotinoides , Oryza , Poluentes do Solo , Oryza/metabolismo , Oryza/crescimento & desenvolvimento , Cádmio/metabolismo , Neonicotinoides/metabolismo , Inseticidas/metabolismo , Poluentes do Solo/metabolismo , Guanidinas/metabolismo , Quelantes , Nitrocompostos/metabolismo , Tiazóis/metabolismo
4.
Ecotoxicol Environ Saf ; 272: 116084, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38350217

RESUMO

Polyhexamethylene guanidine (PHMG) is manufactured and applied extensively due to its superior disinfectant capabilities. However, the inhalatory exposure to PHMG aerosols is increasingly recognized as a potential instigator of pulmonary fibrosis, prompting an urgent call for elucidation of the underlying pathophysiological mechanisms. Within this context, alveolar macrophages play a pivotal role in the primary immune defense in the respiratory tract. Dysregulated lipid metabolism within alveolar macrophages leads to the accumulation of foam cells, a process that is intimately linked with the pathogenesis of pulmonary fibrosis. Therefore, this study examines PHMG's effects on alveolar macrophage foaminess and its underlying mechanisms. We conducted a 3-week inhalation exposure followed by a 3-week recovery period in C57BL/6 J mice using a whole-body exposure system equipped with a disinfection aerosol generator (WESDAG). The presence of lipid-laden alveolar macrophages and downregulation of pulmonary tissue lipid transport proteins ABCA1 and ABCG1 were observed in mice. In cell culture models involving lipid-loaded macrophages, we demonstrated that PHMG promotes foam cell formation by inhibiting lipid efflux in mouse alveolar macrophages. Furthermore, PHMG-induced foam cells were found to promote an increase in the release of TGF-ß1, fibronectin deposition, and collagen remodeling. In vivo interventions were subsequently implemented on mice exposed to PHMG aerosols, aiming to restore macrophage lipid efflux function. Remarkably, this intervention demonstrated the potential to retard the progression of pulmonary fibrosis. In conclusion, this study underscores the pivotal role of macrophage foaming in the pathogenesis of PHMG disinfectants-induced pulmonary fibrosis. Moreover, it provides compelling evidence to suggest that the regulation of macrophage efflux function holds promise for mitigating the progression of pulmonary fibrosis, thereby offering novel insights into the mechanisms underlying inhaled PHMG disinfectants-induced pulmonary fibrosis.


Assuntos
Desinfetantes , Fibrose Pulmonar , Camundongos , Animais , Fibrose Pulmonar/metabolismo , Guanidina/toxicidade , Guanidina/metabolismo , Camundongos Endogâmicos C57BL , Aerossóis e Gotículas Respiratórios , Pulmão , Guanidinas/metabolismo , Macrófagos , Desinfetantes/farmacologia , Lipídeos
5.
Int J Mol Sci ; 24(8)2023 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-37108521

RESUMO

Streptococcus suis, an encapsulated zoonotic pathogen, has been reported to cause a variety of infectious diseases, such as meningitis and streptococcal-toxic-shock-like syndrome. Increasing antimicrobial resistance has triggered the need for new treatments. In the present study, we found that isopropoxy benzene guanidine (IBG) significantly attenuated the effects caused by S. suis infection, in vivo and in vitro, by killing S. suis and reducing S. suis pathogenicity. Further studies showed that IBG disrupted the integrity of S. suis cell membranes and increased the permeability of S. suis cell membranes, leading to an imbalance in proton motive force and the accumulation of intracellular ATP. Meanwhile, IBG antagonized the hemolysis activity of suilysin and decreased the expression of Sly gene. In vivo, IBG improved the viability of S. suis SS3-infected mice by reducing tissue bacterial load. In conclusion, IBG is a promising compound for the treatment of S. suis infections, given its antibacterial and anti-hemolysis activity.


Assuntos
Infecções Estreptocócicas , Streptococcus suis , Animais , Camundongos , Streptococcus suis/genética , Benzeno , Guanidina , Infecções Estreptocócicas/tratamento farmacológico , Infecções Estreptocócicas/microbiologia , Guanidinas/farmacologia , Guanidinas/uso terapêutico , Guanidinas/metabolismo , Proteínas Hemolisinas/metabolismo
6.
Artif Cells Nanomed Biotechnol ; 49(1): 651-661, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34751061

RESUMO

Gusperimus is an anti-inflammatory drug that has shown to be effective in managing autoimmunity and preventing graft rejection. This is unstable and easily broken down into cytotoxic components. We encapsulated gusperimus binding it covalently to squalene obtaining squalene-gusperimus nanoparticles (Sq-GusNPs). These nanoparticles enhanced the immunosuppressive effect of gusperimus in both mouse macrophages and T cells. The half-maximal inhibitory concentration in macrophages was 9-fold lower for Sq-GusNPs compared with the free drug. The anti-inflammatory effect of the Sq-GusNPs was maintained over time without cytotoxicity. By studying nanoparticles uptake by cells with flow cytometry, we demonstrated that Sq-GusNPs are endocytosed by macrophages after binding to low-density lipoprotein receptors (LDLR). In presence of cathepsin B or D release of gusperimus is increased demonstrating the participation of proteases in the release process. Our approach may allow the application of Sq-GusNPs for effective management of inflammatory disorders including autoimmunity and graft rejection.


Assuntos
Nanopartículas , Esqualeno , Animais , Guanidinas/metabolismo , Macrófagos/metabolismo , Camundongos , Esqualeno/metabolismo , Esqualeno/farmacologia
7.
Mol Pharm ; 18(10): 3811-3819, 2021 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-34519204

RESUMO

To develop novel norepinephrine transporter (NET)-targeting positron emission tomography (PET) probes with optimal pharmacokinetic properties, a series of meta-bromobenzylguanidine derivatives was synthesized. 4-Fluorodiethoxyethane-3-bromobenzylguanidine (compound 12) showed relatively good affinity for the NET (IC50 = 1.00 ± 0.04 µM). The corresponding radiotracer 18F-12 was prepared in high radiochemical purity (>98%) via a three-step method. The in vitro cellular uptake results demonstrated that 18F-12 was specifically taken up by NET-expressing SK-N-SH cells by the uptake-1 mechanism. Biodistribution studies in mice showed that 18F-12 exhibited high cardiac uptake (10.45 ± 0.66 %ID/g at 5 min p.i. and 6.44 ± 0.40 %ID/g at 120 min p.i.), faster liver clearance, and a lower dose of absorbed radiation than [123I]-labeled meta-iodobenzylguanidine ([123I]MIBG). Small animal PET imaging confirmed the high heart-to-background ratio of 18F-12 and the uptake-1 mechanism specific for the NET in rats, indicating its potential as a promising PET radiotracer for cardiac sympathetic nerve imaging.


Assuntos
Bromobenzenos/metabolismo , Guanidinas/metabolismo , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/metabolismo , Tomografia por Emissão de Pósitrons , Animais , Bromobenzenos/farmacocinética , Linhagem Celular Tumoral , Radioisótopos de Flúor/farmacocinética , Guanidinas/farmacocinética , Humanos , Camundongos Endogâmicos ICR , Tomografia por Emissão de Pósitrons/métodos
8.
J Virol ; 95(19): e0086121, 2021 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-34160253

RESUMO

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.


Assuntos
Antivirais/farmacologia , Ésteres/farmacologia , Guanidinas/farmacologia , SARS-CoV-2/efeitos dos fármacos , Serina Endopeptidases/química , Serina Endopeptidases/farmacologia , Serina Proteases/farmacologia , Antivirais/química , COVID-19/prevenção & controle , Carcinoma de Células Escamosas , Ésteres/química , Ésteres/metabolismo , Guanidinas/química , Guanidinas/metabolismo , Humanos , Simulação de Dinâmica Molecular , Neoplasias Bucais , Domínios Proteicos , Alinhamento de Sequência , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Serina Proteases/química , Serina Proteases/metabolismo , Inibidores de Serina Proteinase/química , Inibidores de Serina Proteinase/farmacologia , Internalização do Vírus/efeitos dos fármacos , Tratamento Farmacológico da COVID-19
9.
Chem Biol Interact ; 338: 109428, 2021 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-33647240

RESUMO

Camostat mesylate, a potent inhibitor of the human transmembrane protease, serine 2 (TMPRSS2), is currently under investigation for its effectiveness in COVID-19 patients. For its safe application, the risks of camostat mesylate to induce pharmacokinetic drug-drug interactions with co-administered drugs should be known. We therefore tested in vitro the potential inhibition of important efflux (P-glycoprotein (P-gp, ABCB1), breast cancer resistance protein (BCRP, ABCG2)), and uptake transporters (organic anion transporting polypeptides OATP1B1, OATP1B3, OATP2B1) by camostat mesylate and its active metabolite 4-(4-guanidinobenzoyloxy)phenylacetic acid (GBPA). Transporter inhibition was evaluated using fluorescent probe substrates in transporter over-expressing cell lines and compared to the respective parental cell lines. Moreover, possible mRNA induction of pharmacokinetically relevant genes regulated by the nuclear pregnane X receptor (PXR) and aryl hydrocarbon receptor (AhR) was analysed in LS180 cells by quantitative real-time PCR. The results of our study for the first time demonstrated that camostat mesylate and GBPA do not relevantly inhibit P-gp, BCRP, OATP1B1 or OATP1B3. Only OATP2B1 was profoundly inhibited by GBPA with an IC50 of 11 µM. Induction experiments in LS180 cells excluded induction of PXR-regulated genes such as cytochrome P450 3A4 (CYP3A4) and ABCB1 and AhR-regulated genes such as CYP1A1 and CYP1A2 by camostat mesylate and GBPA. Together with the summary of product characteristics of camostat mesylate indicating no inhibition of CYP1A2, 2C9, 2C19, 2D6, and 3A4 in vitro, our data suggest a low potential of camostat mesylate to act as a perpetrator in pharmacokinetic drug-drug interactions. Only inhibition of OATP2B1 by GBPA warrants further investigation.


Assuntos
Interações Medicamentosas , Ésteres/metabolismo , Guanidinas/metabolismo , Inibidores de Serina Proteinase/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Ésteres/química , Ésteres/farmacologia , Guanidinas/química , Guanidinas/farmacologia , Humanos , Transportadores de Ânions Orgânicos/antagonistas & inibidores , Transportadores de Ânions Orgânicos/metabolismo , Receptor de Pregnano X/genética , Receptor de Pregnano X/metabolismo , RNA Mensageiro/metabolismo , Receptores de Hidrocarboneto Arílico/genética , Receptores de Hidrocarboneto Arílico/metabolismo , Serina Endopeptidases/química , Serina Endopeptidases/metabolismo , Inibidores de Serina Proteinase/química , Inibidores de Serina Proteinase/farmacologia
10.
Bioconjug Chem ; 31(10): 2446-2455, 2020 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-32991164

RESUMO

Benzylguanidine, a small cationic and amphiphilic molecule, exhibits a high affinity to C-X-C chemokine receptor type 4 (CXCR 4) and a membrane penetration ability. It has not been used as a functional moiety of nanocarriers for the systemic delivery of chemotherapeutic drugs in tumor therapy. In this study, we investigated the membrane penetration of benzylguanidine-conjugated nanocarriers and their efficiency and safety for targeted delivery of doxorubicin (DOX) in CXCR 4 positive tumors. We conjugated the benzylguanidine bearing guanidinobenzoic acid onto the cystamine bismethacrylamide cross-linked chitosan-poly(methyl methacrylate) nanoparticles, which were then decorated with lactobionic acid (abbreviated as LGCC NPs). A small proportion of LGCC NPs were able to directly penetrate the plasma membrane to enter cells, thereby circumventing endocytic vesicles. The DOX-loaded LGCC NPs (LGCC NPs/DOX) displayed good stability under extracellular physiological conditions and reduction-triggered drug release under high glutathione (GSH) concentration. Moreover, LGCC NPs/DOX showed an increase in tumor-targeted cellular uptake through receptor-mediated endocytosis, enhanced endo/lysosomal escape, and a high nuclear distribution. More importantly, LGCC NPs/DOX significantly suppressed the in vitro and in vivo proliferation of CXCR 4 positive hepatocarcinoma and breast cancer. The findings provide a guideline for the combined application of benzylguanidine and other functional groups in antitumor nanomedicines.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Doxorrubicina/administração & dosagem , Portadores de Fármacos/química , Galactose/análogos & derivados , Guanidinas/química , Neoplasias/tratamento farmacológico , Receptores CXCR4/metabolismo , Animais , Antibióticos Antineoplásicos/farmacocinética , Antibióticos Antineoplásicos/farmacologia , Antibióticos Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Quitosana/análogos & derivados , Quitosana/metabolismo , Doxorrubicina/farmacocinética , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Portadores de Fármacos/metabolismo , Sistemas de Liberação de Medicamentos , Galactose/metabolismo , Guanidinas/metabolismo , Humanos , Camundongos , Nanopartículas/química , Nanopartículas/metabolismo , Neoplasias/metabolismo
11.
BMC Cancer ; 20(1): 724, 2020 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-32758183

RESUMO

BACKGROUND: Breast tumor initiating cells (BTIC) are stem-like cells that initiate and sustain tumor growth, and drive disease recurrence. Identifying therapies targeting BTIC has been hindered due primarily to their scarcity in tumors. We previously reported that BTIC frequency ranges between 15% and 50% in multiple mammary tumors of 3 different transgenic mouse models of breast cancer and that this frequency is maintained in tumor cell populations cultured in serum-free, chemically defined media as non-adherent tumorspheres. The latter enabled high-throughput screening of small molecules for their capacity to affect BTIC survival. Antagonists of several serotonin receptors (5-HTRs) were among the hit compounds. The most potent compound we identified, SB-699551, selectively binds to 5-HT5A, a Gαi/o protein coupled receptor (GPCR). METHODS: We evaluated the activity of structurally unrelated selective 5-HT5A antagonists using multiple orthogonal assays of BTIC frequency. Thereafter we used a phosphoproteomic approach to uncover the mechanism of action of SB-699551. To validate the molecular target of the antagonists, we used the CRISPR-Cas9 gene editing technology to conditionally knockout HTR5A in a breast tumor cell line. RESULTS: We found that selective antagonists of 5-HT5A reduced the frequency of tumorsphere initiating cells residing in breast tumor cell lines and those of patient-derived xenografts (PDXs) that we established. The most potent compound among those tested, SB-699551, reduced the frequency of BTIC in ex vivo assays and acted in concert with chemotherapy to shrink human breast tumor xenografts in vivo. Our phosphoproteomic experiments established that exposure of breast tumor cells to SB-699551 elicited signaling changes in the canonical Gαi/o-coupled pathway and the phosphoinositide 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) axis. Moreover, conditional mutation of the HTR5A gene resulted in the loss of tumorsphere initiating cells and BTIC thus mimicking the effect of SB-699551. CONCLUSIONS: Our data provide genetic, pharmacological and phosphoproteomic evidence consistent with the on-target activity of SB-699551. The use of such agents in combination with cytotoxic chemotherapy provides a novel therapeutic approach to treat breast cancer.


Assuntos
Compostos de Bifenilo/farmacologia , Neoplasias da Mama/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Receptores de Serotonina/metabolismo , Antagonistas da Serotonina/farmacologia , Animais , Antineoplásicos/farmacologia , Compostos de Bifenilo/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Classe I de Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Feminino , Técnicas de Inativação de Genes , Guanidinas/química , Guanidinas/metabolismo , Guanidinas/farmacologia , Xenoenxertos , Humanos , Isoquinolinas/química , Isoquinolinas/metabolismo , Isoquinolinas/farmacologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Transplante de Neoplasias , Proteômica , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Serotonina/genética , Antagonistas da Serotonina/química , Antagonistas da Serotonina/metabolismo
12.
Proc Natl Acad Sci U S A ; 117(24): 13490-13498, 2020 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-32461356

RESUMO

The voltage-gated Hv1 proton channel is a ubiquitous membrane protein that has roles in a variety of cellular processes, including proton extrusion, pH regulation, production of reactive oxygen species, proliferation of cancer cells, and increased brain damage during ischemic stroke. A crystal structure of an Hv1 construct in a putative closed state has been reported, and structural models for the channel open state have been proposed, but a complete characterization of the Hv1 conformational dynamics under an applied membrane potential has been elusive. We report structural models of the Hv1 voltage-sensing domain (VSD), both in a hyperpolarized state and a depolarized state resulting from voltage-dependent conformational changes during a 10-µs-timescale atomistic molecular dynamics simulation in an explicit membrane environment. In response to a depolarizing membrane potential, the S4 helix undergoes an outward displacement, leading to changes in the VSD internal salt-bridge network, resulting in a reshaping of the permeation pathway and a significant increase in hydrogen bond connectivity throughout the channel. The total gating charge displacement associated with this transition is consistent with experimental estimates. Molecular docking calculations confirm the proposed mechanism for the inhibitory action of 2-guanidinobenzimidazole (2GBI) derived from electrophysiological measurements and mutagenesis. The depolarized structural model is also consistent with the formation of a metal bridge between residues located in the core of the VSD. Taken together, our results suggest that these structural models are representative of the closed and open states of the Hv1 channel.


Assuntos
Ativação do Canal Iônico , Canais Iônicos/química , Canais Iônicos/metabolismo , Cristalografia por Raios X , Guanidinas/metabolismo , Humanos , Ligação de Hidrogênio , Canais Iônicos/genética , Potenciais da Membrana , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Mutação , Conformação Proteica , Prótons
13.
Proc Natl Acad Sci U S A ; 117(17): 9338-9348, 2020 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-32284409

RESUMO

Oxidation of guanine generates several types of DNA lesions, such as 8-oxoguanine (8OG), 5-guanidinohydantoin (Gh), and spiroiminodihydantoin (Sp). These guanine-derived oxidative DNA lesions interfere with both replication and transcription. However, the molecular mechanism of transcription processing of Gh and Sp remains unknown. In this study, by combining biochemical and structural analysis, we revealed distinct transcriptional processing of these chemically related oxidized lesions: 8OG allows both error-free and error-prone bypass, whereas Gh or Sp causes strong stalling and only allows slow error-prone incorporation of purines. Our structural studies provide snapshots of how polymerase II (Pol II) is stalled by a nonbulky Gh lesion in a stepwise manner, including the initial lesion encounter, ATP binding, ATP incorporation, jammed translocation, and arrested states. We show that while Gh can form hydrogen bonds with adenosine monophosphate (AMP) during incorporation, this base pair hydrogen bonding is not sufficient to hold an ATP substrate in the addition site and is not stable during Pol II translocation after the chemistry step. Intriguingly, we reveal a unique structural reconfiguration of the Gh lesion in which the hydantoin ring rotates ∼90° and is perpendicular to the upstream base pair planes. The perpendicular hydantoin ring of Gh is stabilized by noncanonical lone pair-π and CH-π interactions, as well as hydrogen bonds. As a result, the Gh lesion, as a functional mimic of a 1,2-intrastrand crosslink, occupies canonical -1 and +1 template positions and compromises the loading of the downstream template base. Furthermore, we suggest Gh and Sp lesions are potential targets of transcription-coupled repair.


Assuntos
Guanidinas/química , Guanosina/análogos & derivados , Hidantoínas/química , RNA Polimerase II/metabolismo , Compostos de Espiro/química , Pareamento de Bases , DNA/química , DNA/metabolismo , Dano ao DNA/fisiologia , Reparo do DNA/fisiologia , Guanidinas/metabolismo , Guanina/metabolismo , Guanosina/química , Guanosina/metabolismo , Hidantoínas/metabolismo , Oxirredução , Estresse Oxidativo/fisiologia , Purinas/metabolismo , RNA Polimerase II/fisiologia , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Compostos de Espiro/metabolismo , Transcrição Gênica/fisiologia , Ativação Transcricional/fisiologia
14.
Chem Biol Interact ; 324: 109087, 2020 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-32294457

RESUMO

Despite advances in cancer treatment modalities, DNA still stands as one of the targets for anticancer agents. DNA minor groove binders (MGBs) represent an important investigational chemotherapeutic class with promising cytotoxic capacity. Herein this study reports the potent cytotoxic effect of a series of repurposed flexible bis-imidamides 1-4, triaryl bis-guanidine 5 and bis-N-substituted guanidines 6,7 having a 1,4-diphenoxybenzene scaffold backbone on MCF-7 and MDA-MB-231 breast cancer cell lines. Of these compounds, imidamide 4 was chosen for further in-vitro, in-vivo and molecular dynamics (MD) studies owing to its promising anti-tumor activity, with IC50 values on MCF-7 and MDA-MB-231 breast cancer cell lines of 1.9 and 2.08 µM, respectively. Annexin V/propidium iodide apoptosis assay revealed apoptosis induction on imidamide 4 treated MCF-7 cells. RT-PCR assay results demonstrated the proapoptotic effect of compound 4 through increase of mRNA levels of the pro-apoptotic genes; p53, PUMA, and Bax, and inhibiting the anti-apoptotic Bcl-2 gene expression in MCF-7 cells. Moreover, compound 4 induced a G0/G1 cell-cycle arrest in MCF-7 in a dose-dependent manner. Corroborating in-vivo experiments on Ehrlich ascites carcinoma (EAC)-bearing mice, reflected the anticancer strength of derivative 4. For further target validation, molecular dynamics (MD) studies demonstrated an energetically favorable binding of imidamide 4 with the DNA minor groove AT rich site. In effect, imidamide 4 can be viewed as a promising hit dicationic compound with good cytotoxic and apoptotic inducing activity against breast cancer that can be adopted for future optimization.


Assuntos
Antineoplásicos/uso terapêutico , Antioxidantes/uso terapêutico , Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , DNA/metabolismo , Guanidinas/uso terapêutico , Animais , Antineoplásicos/química , Antineoplásicos/metabolismo , Antioxidantes/química , Antioxidantes/metabolismo , Carcinoma de Ehrlich/tratamento farmacológico , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Guanidinas/química , Guanidinas/metabolismo , Humanos , Fígado/patologia , Camundongos , Simulação de Dinâmica Molecular , Estrutura Molecular , Relação Estrutura-Atividade
15.
J Plant Physiol ; 246-247: 153092, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32065919

RESUMO

The mechanisms regulating, and modulating potato wound-healing processes are of great importance in reducing tuber infections, reducing shrinkage and maintaining quality and nutritional value for growers and consumers. Wound-induced changes in tuber polyamine metabolism have been linked to the modulation of wound healing (WH) and in possibly providing the crucial amount of H2O2 required for suberization processes. In this investigation we determined the effect of inhibition of specific steps within the pathway of polyamine metabolism on polyamine content and the initial accumulation of suberin polyphenolics (SPP) during WH. The accumulation of SPP represents a critical part of the beginning or inchoate phase of tuber WH during closing-layer formation because it serves as a barrier to bacterial infection and is a requisite for the accumulation of suberin polyaliphatics which provide the barrier to fungal infection. Results showed that the inhibitor treatments that caused changes in polyamine content generally did not influence wound-induced accumulation of SPP. Such lack of correlation was found for inhibitors involved in metabolism and oxidation of putrescine (arginine decarboxylase, ornithine decarboxylase, and diamine oxidase). However, accumulation of SPP was dramatically reduced by treatment with guazatine, a potent inhibitor of polyamine oxidase (PAO), and methylglyoxal-bis(guanylhydrazone), a putative inhibitor of S-adenosylmethione decarboxylase which may also cross-react to inhibit PAO. The mode of action of these inhibitors is presumed to be blockage of essential H2O2 production within the WH cell wall. These results are of great importance in understanding the mechanisms modulating WH and ultimately controlling related infections and associated postharvest losses.


Assuntos
Diaminas/antagonistas & inibidores , Lipídeos/biossíntese , Proteínas de Plantas/metabolismo , Tubérculos/metabolismo , Poliaminas/antagonistas & inibidores , Solanum tuberosum/metabolismo , Carboxiliases/metabolismo , Diaminas/metabolismo , Guanidinas/metabolismo , Mitoguazona/metabolismo , Oxirredução , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/metabolismo , Poliaminas/metabolismo , Putrescina/metabolismo , Solanum tuberosum/enzimologia , Poliamina Oxidase
16.
J Med Chem ; 63(5): 2051-2073, 2020 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-31268317

RESUMO

High risk neuroblastoma often recurs, even with aggressive treatments. Clinical evidence suggests that proliferative activities are predictive of poor outcomes. This report describes syntheses, characterization, and biological properties of theranostic guanidines that target norepinephrine transporter and undergo intracellular processing, and subsequently their catabolites are efficiently incorporated into DNA of proliferating neuroblastoma cells. Radioactive guanidines are synthesized from 5-radioiodo-2'-deoxyuridine, a molecular radiotherapy platform with clinically proven minimal toxicities and DNA-targeting properties. The transport of radioactive guanidines into neuroblastoma cells is active as indicated by the competitive suppression of cellular uptake by meta-iodobenzylguanidine. The rate of intracellular processing and DNA uptake is influenced by the agent's catabolic stability and cell population doubling times. The radiotoxicity is directly proportional to DNA uptake and duration of exposure. Biodistribution of 5-[125I]iodo-3'-O-(ε-guanidinohexanoyl)-2'-deoxyuridine in a mouse neuroblastoma model shows significant tumor retention of radioactivity. Neuroblastoma xenografts regress in response to the clinically achievable doses of this agent.


Assuntos
DNA/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Guanidinas/metabolismo , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/metabolismo , Nanomedicina Teranóstica/métodos , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Feminino , Guanidinas/administração & dosagem , Humanos , Masculino , Camundongos , Camundongos Nus , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
17.
Cells ; 9(1)2019 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-31878359

RESUMO

Polyhexamethylene guanidine phosphate (PHMG-p), a member of the polymeric guanidine family, has strong antimicrobial activity and may increase the risk of inflammation-associated pulmonary fibrosis. However, the effect of PHMG-p on the barrier function of the bronchial epithelium is unknown. Epithelial barrier functioning is maintained by tight junctions (TJs); damage to these TJs is the major cause of epithelial barrier breakdown during lung inflammation. The present study showed that, in BEAS-2B human bronchial epithelial cells, exposure to PHMG-p reduced the number of TJs and the E-cadherin level and impaired the integrity of the F-actin architecture. Furthermore, exposure to PHMG-p stimulated the calcium-dependent protease calpain-1, which breaks down TJs. However, treatment with the calpain-1 inhibitor, ALLN, reversed the PHMG-p-mediated impairment of TJs and the F-actin architecture. Furthermore, exposure to PHMG-p increased the intracellular Ca2+ level via P2X purinoreceptor 7 (P2RX7) and inhibition of P2RX7 abolished the PHMG-p-induced calpain-1 activity and protein degradation and increased the intracellular Ca2+ level. Although exposure to PHMG-p increased the extracellular ATP level, hydrolysis of extracellular ATP by apyrase did not influence its detrimental effect on bronchial epithelial cells. These results implicate the impairment of TJs and the F-actin architecture in the pathogenesis of pulmonary diseases.


Assuntos
Guanidinas/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Junções Íntimas/metabolismo , Actinas/metabolismo , Antígenos CD/metabolismo , Apirase/metabolismo , Caderinas/metabolismo , Cálcio/metabolismo , Calpaína/metabolismo , Linhagem Celular , Células Epiteliais/efeitos dos fármacos , Humanos , Pulmão/metabolismo , Fibrose Pulmonar/metabolismo , Transdução de Sinais/efeitos dos fármacos
18.
Toxicol Appl Pharmacol ; 383: 114777, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31626844

RESUMO

Neonicotinoids are one of most widely used pesticides targeting nicotinic acetylcholine receptors (nAChRs) of insects. Recent epidemiological evidence revealed increasing amounts of neonicotinoids detected in human samples, raising the critical question of whether neonicotinoids affect human health. We investigated the effects of a neonicotinoid pesticide clothianidin (CTD) on human neuroblastoma SH-SY5Y cells as in vitro models of human neuronal cells. Cellular and functional effects of micromolar doses of CTD were evaluated by changes in cell growth, intracellular signaling activities and gene expression profiles. We examined further the effects of CTD on neuronal differentiation by measuring neurite outgrowth. Exposure to CTD (1-100 µM) significantly increased the number of cells within 24 h of culture. The nAChRs antagonists, mecamylamine and SR16584, inhibited this effect, suggesting human α3ß4 nAChRs could be targets of neonicotinoids. We observed a transient intracellular calcium influx and increased phosphorylation of extracellular signal-regulated kinase 1/2 shortly after exposure to CTD. Transcriptome analysis revealed that CTD down-regulated genes involved in neuronal function (e.g., formation of filopodia and calcium ion influx) and morphology (e.g., axon guidance signaling and cytoskeleton signaling); these changes were reflected by a finding of increased neurite length during neuronal differentiation. These findings provide novel insight into the potential risks of neonicotinoids to the human nervous system.


Assuntos
Proliferação de Células/efeitos dos fármacos , Guanidinas/toxicidade , Neonicotinoides/toxicidade , Neuritos/efeitos dos fármacos , Praguicidas/toxicidade , Tiazóis/toxicidade , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Relação Dose-Resposta a Droga , Redes Reguladoras de Genes/efeitos dos fármacos , Redes Reguladoras de Genes/fisiologia , Guanidinas/metabolismo , Humanos , Neonicotinoides/metabolismo , Neuritos/metabolismo , Neuritos/patologia , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Praguicidas/metabolismo , Receptores Nicotínicos/metabolismo , Tiazóis/metabolismo
19.
J Med Chem ; 62(15): 6985-6991, 2019 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-31283215

RESUMO

Meta-iodobenzylguanidine (MIBG) is a ligand with high affinity against norepinephrine transporter (NET) that has been used for diagnostic imaging and radionuclide therapy of NET-expressing tumors, such as neuroblastoma. We hypothesize that MIBG can be used as a ligand for development of new anticancer drugs targeting NET-expressing neuroblastoma (NB). To test our hypothesis, we synthesized two MIBG-based anticancer copper complexes [Cu(m-TSBG)2 and Cu(p-TSBG)2] by conjugation of a thiosemicarbazone copper group onto MIBG ligand. Both Cu(m-TSBG)2 and Cu(p-TSBG)2 compounds showed potent anticancer activity against NB cells (BE2C and SK-N-DZ cells). The NB-specific anticancer activity of Cu(m-TSBG)2 and Cu(p-TSBG)2 was further demonstrated by the reduced anticancer activities when nonconjugated MIBG ligand was used to competitively block binding of Cu(m-TSBG)2 or Cu(p-TSBG)2 onto NET-expressing NB cells. Both Cu(m-TSBG)2 or Cu(p-TSBG)2 compounds hold potential as promising new drugs for targeted therapy of neuroblastoma and other NET-expressing tumors.


Assuntos
3-Iodobenzilguanidina/metabolismo , Antineoplásicos/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Neuroblastoma/metabolismo , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/metabolismo , Semicarbazidas/metabolismo , 3-Iodobenzilguanidina/química , 3-Iodobenzilguanidina/farmacologia , Antineoplásicos/administração & dosagem , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Cobre/química , Cobre/metabolismo , Cobre/farmacologia , Guanidinas/química , Guanidinas/metabolismo , Guanidinas/farmacologia , Humanos , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/antagonistas & inibidores , Semicarbazidas/química , Semicarbazidas/farmacologia
20.
Environ Pollut ; 252(Pt A): 856-862, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31202138

RESUMO

Neonicotinoid insecticides have been widely used throughout the world over the last two decades. In the present study, we investigated the degradation of neonicotinoid insecticides nitenpyram (NIT) and dinotefuran (DIN) by the white-rot fungus Phanerochaete sordida YK-624. While NIT was completely degraded by P. sordida YK-624 under ligninolytic conditions, only a 20% decrease was observed under nonligninolytic conditions. On the other hand, P. sordida YK-624 degraded 31% of DIN under ligninolytic conditions after a 20-day incubation, while it did not degrade DIN under nonligninolytic conditions. We found that cytochromes P450 played a key role in the biotransformation of NIT and DIN by P. sordida YK-624. A novel NIT metabolite (E)-N-((6-chloropyridin-3-yl)methyl)-N-ethyl-N'-hydroxy acetimidamide (CPMHA) and a novel DIN metabolite N-((4aS,7aS,E)-1-methylhexahydrofuro[2,3-d]pyrimidin-2(1H)-ylidene)nitramide (PHPF) were identified in this study. In addition, to evaluate neurotoxicity, the effects of NIT, DIN and their metabolites on the viability of human neuroblastoma cells SH-SY5Y were determined. PHPF showed higher neurological toxicity than DIN, whereas the metabolite of NIT, CPMHA, showed no toxic effect. Our results indicated that the neurological toxicity of NIT could be effectively removed by P. sordida YK-624.


Assuntos
Biodegradação Ambiental , Guanidinas/metabolismo , Inativação Metabólica/fisiologia , Inseticidas/metabolismo , Neonicotinoides/metabolismo , Nitrocompostos/metabolismo , Phanerochaete/metabolismo , Linhagem Celular Tumoral , Sistema Enzimático do Citocromo P-450/metabolismo , Humanos , Neurotoxinas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA