Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 218
Filtrar
1.
Mar Drugs ; 22(5)2024 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-38786608

RESUMO

We identified a new human voltage-gated potassium channel blocker, NnK-1, in the jellyfish Nemopilema nomurai based on its genomic information. The gene sequence encoding NnK-1 contains 5408 base pairs, with five introns and six exons. The coding sequence of the NnK-1 precursor is 894 nucleotides long and encodes 297 amino acids containing five presumptive ShK-like peptides. An electrophysiological assay demonstrated that the fifth peptide, NnK-1, which was chemically synthesized, is an effective blocker of hKv1.3, hKv1.4, and hKv1.5. Multiple-sequence alignment with cnidarian Shk-like peptides, which have Kv1.3-blocking activity, revealed that three residues (3Asp, 25Lys, and 34Thr) of NnK-1, together with six cysteine residues, were conserved. Therefore, we hypothesized that these three residues are crucial for the binding of the toxin to voltage-gated potassium channels. This notion was confirmed by an electrophysiological assay with a synthetic peptide (NnK-1 mu) where these three peptides were substituted with 3Glu, 25Arg, and 34Met. In conclusion, we successfully identified and characterized a new voltage-gated potassium channel blocker in jellyfish that interacts with three different voltage-gated potassium channels. A peptide that interacts with multiple voltage-gated potassium channels has many therapeutic applications in various physiological and pathophysiological contexts.


Assuntos
Peptídeos , Bloqueadores dos Canais de Potássio , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Cifozoários , Animais , Humanos , Bloqueadores dos Canais de Potássio/farmacologia , Bloqueadores dos Canais de Potássio/química , Canais de Potássio de Abertura Dependente da Tensão da Membrana/antagonistas & inibidores , Peptídeos/farmacologia , Peptídeos/química , Sequência de Aminoácidos , Venenos de Cnidários/farmacologia , Venenos de Cnidários/química , Alinhamento de Sequência
2.
J Chem Inf Model ; 63(10): 3043-3053, 2023 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-37143234

RESUMO

Peptide toxins that adopt the ShK fold can inhibit the voltage-gated potassium channel KV1.3 with IC50 values in the pM range and are therefore potential leads for drugs targeting autoimmune and neuroinflammatory diseases. Nuclear magnetic resonance (NMR) relaxation measurements and pressure-dependent NMR have shown that, despite being cross-linked by disulfide bonds, ShK itself is flexible in solution. This flexibility affects the local structure around the pharmacophore for the KV1.3 channel blockade and, in particular, the relative orientation of the key Lys and Tyr side chains (Lys22 and Tyr23 in ShK) and has implications for the design of KV1.3 inhibitors. In this study, we have performed molecular dynamics (MD) simulations on ShK and a close homologue, HmK, to probe the conformational space occupied by the Lys and Tyr residues, and docked the different conformations with a recently determined cryo-EM structure of the KV1.3 channel. Although ShK and HmK have 60% sequence identity, their dynamic behaviors are quite different, with ShK sampling a broad range of conformations over the course of a 5 µs MD simulation, while HmK is relatively rigid. We also investigated the importance of conformational dynamics, in particular the distance between the side chains of the key dyad Lys22 and Tyr23, for binding to KV1.3. Although these peptides have quite different dynamics, the dyad in both adopts a similar configuration upon binding, revealing a conformational selection upon binding to KV1.3 in the case of ShK. Both peptides bind to KV1.3 with Lys22 occupying the pore of the channel. Intriguingly, the more flexible peptide, ShK, binds with significantly higher affinity than HmK.


Assuntos
Venenos de Cnidários , Anêmonas-do-Mar , Animais , Canal de Potássio Kv1.3/química , Canal de Potássio Kv1.3/metabolismo , Venenos de Cnidários/química , Venenos de Cnidários/metabolismo , Venenos de Cnidários/farmacologia , Anêmonas-do-Mar/química , Anêmonas-do-Mar/metabolismo , Peptídeos/química , Conformação Molecular , Bloqueadores dos Canais de Potássio/farmacologia , Bloqueadores dos Canais de Potássio/química , Canal de Potássio Kv1.2/metabolismo
3.
Mar Drugs ; 21(3)2023 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-36976217

RESUMO

Nowadays, major attention is being paid to curing different types of cancers and is focused on natural resources, including oceans and marine environments. Jellyfish are marine animals with the ability to utilize their venom in order to both feed and defend. Prior studies have displayed the anticancer capabilities of various jellyfish. Hence, we examined the anticancer features of the venom of Cassiopea andromeda and Catostylus mosaicus in an in vitro situation against the human pulmonary adenocarcinoma (A549) cancer cell line. The MTT assay demonstrated that both mentioned venoms have anti-tumoral ability in a dose-dependent manner. Western blot analysis proved that both venoms can increase some pro-apoptotic factors and reduce some anti-apoptotic molecules that lead to the inducing of apoptosis in A549 cells. GC/MS analysis demonstrated some compounds with biological effects, including anti-inflammatory, antioxidant and anti-cancer activities. Molecular docking and molecular dynamic showed the best position of each biologically active component on the different death receptors, which are involved in the process of apoptosis in A549 cells. Ultimately, this study has proven that both venoms of C. andromeda and C. mosaicus have the capability to suppress A549 cells in an in vitro condition and they might be utilized in order to design and develop brand new anticancer agents in the near future.


Assuntos
Adenocarcinoma , Cnidários , Venenos de Cnidários , Neoplasias Pulmonares , Cifozoários , Animais , Humanos , Venenos de Cnidários/farmacologia , Venenos de Cnidários/química , Células A549 , Simulação de Acoplamento Molecular , Adenocarcinoma/tratamento farmacológico , Apoptose , Neoplasias Pulmonares/tratamento farmacológico
4.
Int J Mol Sci ; 24(5)2023 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-36901790

RESUMO

Infections caused by multi-drug-resistant (MDR) bacteria are a global threat to human health. As venoms are the source of biochemically diverse bioactive proteins and peptides, we investigated the antimicrobial activity and murine skin infection model-based wound healing efficacy of a 13 kDa protein. The active component PaTx-II was isolated from the venom of Pseudechis australis (Australian King Brown or Mulga Snake). PaTx-II inhibited the growth of Gram-positive bacteria in vitro, with moderate potency (MICs of 25 µM) observed against S. aureus, E. aerogenes, and P. vulgaris. The antibiotic activity of PaTx-II was associated with the disruption of membrane integrity, pore formation, and lysis of bacterial cells, as evidenced by scanning and transmission microscopy. However, these effects were not observed with mammalian cells, and PaTx-II exhibited minimal cytotoxicity (CC50 > 1000 µM) toward skin/lung cells. Antimicrobial efficacy was then determined using a murine model of S. aureus skin infection. Topical application of PaTx-II (0.5 mg/kg) cleared S. aureus with concomitant increased vascularization and re-epithelialization, promoting wound healing. As small proteins and peptides can possess immunomodulatory effects to enhance microbial clearance, cytokines and collagen from the wound tissue samples were analyzed by immunoblots and immunoassays. The amounts of type I collagen in PaTx-II-treated sites were elevated compared to the vehicle controls, suggesting a potential role for collagen in facilitating the maturation of the dermal matrix during wound healing. Levels of the proinflammatory cytokines interleukin-1ß (IL-1ß), interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α), cyclooxygenase-2 (COX-2) and interleukin-10 (IL-10), factors known to promote neovascularization, were substantially reduced by PaTx-II treatment. Further studies that characterize the contributions towards efficacy imparted by in vitro antimicrobial and immunomodulatory activity with PaTx-II are warranted.


Assuntos
Anti-Infecciosos , Venenos de Cnidários , Colubridae , Humanos , Animais , Camundongos , Staphylococcus aureus , Austrália , Cicatrização , Anti-Infecciosos/farmacologia , Venenos de Cnidários/farmacologia , Colágeno/farmacologia , Peptídeos/farmacologia , Citocinas/farmacologia , Mamíferos
5.
Molecules ; 26(17)2021 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-34500621

RESUMO

Jellyfish venom is a rich source of bioactive proteins and peptides with various biological activities including antioxidant, antimicrobial and antitumor effects. However, the anti-proliferative activity of the crude extract of Rhopilema nomadica jellyfish venom has not been examined yet. The present study aimed at the investigation of the in vitro effect of R. nomadica venom on liver cancer cells (HepG2), breast cancer cells (MDA-MB231), human normal fibroblast (HFB4), and human normal lung cells (WI-38) proliferation by using MTT assay. The apoptotic cell death in HepG2 cells was investigated using Annexin V-FITC/PI double staining-based flow cytometry analysis, western blot analysis, and DNA fragmentation assays. R. nomadica venom displayed significant dose-dependent cytotoxicity on HepG2 cells after 48 h of treatment with IC50 value of 50 µg/mL and higher toxicity (3:5-fold change) against MDA-MB231, HFB4, and WI-38 cells. R. nomadica venom showed a prominent increase of apoptosis as revealed by cell cycle arrest at G2/M phase, upregulation of p53, BAX, and caspase-3 proteins, and the down-regulation of anti-apoptotic Bcl-2 protein and DNA fragmentation. These findings suggest that R. nomadica venom induces apoptosis in hepatocellular carcinoma cells. To the best of the authors' knowledge, this is the first scientific evidence demonstrating the induction of apoptosis and cell cycle arrest of R. nomadica jellyfish venom.


Assuntos
Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/tratamento farmacológico , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Venenos de Cnidários/farmacologia , Neoplasias Hepáticas/tratamento farmacológico , Cifozoários/metabolismo , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo
6.
Mar Drugs ; 19(1)2021 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-33477357

RESUMO

Arthritis is a widespread inflammatory disease associated with progressive articular surface degradation, ongoing pain, and hyperalgesia causing the development of functional limitations and disability. TRPV1 channel is one of the high-potential targets for the treatment of inflammatory diseases. Polypeptide APHC3 from sea anemone Heteractis crispa is a mode-selective TRPV1 antagonist that causes mild hypothermia and shows significant anti-inflammatory and analgesic activity in different models of pain. We evaluated the anti-inflammatory properties of APHC3 in models of monosodium iodoacetate (MIA)-induced osteoarthritis and complete Freund's adjuvant (CFA)-induced rheumatoid monoarthritis in comparison with commonly used non-steroidal anti-inflammatory drugs (NSAIDs) such as diclofenac, ibuprofen, and meloxicam. Subcutaneous administration of APHC3 (0.1 mg/kg) significantly reversed joint swelling, disability, grip strength impairment, and thermal and mechanical hypersensitivity. The effect of APHC3 was equal to or better than that of reference NSAIDs. Protracted treatment with APHC3 decreased IL-1b concentration in synovial fluid, reduced inflammatory changes in joints, and prevented the progression of cartilage degradation. Therefore, polypeptide APHC3 has the potential to be an analgesic and anti-inflammatory substance for the alleviation of arthritis symptoms.


Assuntos
Analgésicos/farmacologia , Anti-Inflamatórios/farmacologia , Artrite Experimental/tratamento farmacológico , Venenos de Cnidários/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Analgésicos/isolamento & purificação , Animais , Anti-Inflamatórios/isolamento & purificação , Anti-Inflamatórios não Esteroides/farmacologia , Artrite Experimental/fisiopatologia , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/fisiopatologia , Venenos de Cnidários/isolamento & purificação , Modelos Animais de Doenças , Progressão da Doença , Peptídeos e Proteínas de Sinalização Intercelular/isolamento & purificação , Masculino , Osteoartrite/tratamento farmacológico , Osteoartrite/fisiopatologia , Dor/tratamento farmacológico , Dor/fisiopatologia , Ratos , Ratos Sprague-Dawley , Canais de Cátion TRPV/antagonistas & inibidores
7.
ACS Appl Bio Mater ; 4(4): 3360-3373, 2021 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-35014421

RESUMO

Creation of surfaces resistant to the formation of microbial biofilms via biomimicry has been heralded as a promising strategy to protect a range of different materials ranging from boat hulls to medical devices and surgical instruments. In our current study, we describe the successful transfer of a highly effective natural marine biofilm inhibitor to the 2D surface format. A series of cyclic peptides inspired by the natural equinatoxin II protein produced by Beadlet anemone (Actinia equine) have been evaluated for their ability to inhibit the formation of a mixed marine microbial consortium on polyamide reverse osmosis membranes. In solution, the peptides are shown to effectively inhibit settlement and biofilm formation in a nontoxic manner down to 1 nM concentrations. In addition, our study also illustrates how the peptides can be applied to disperse already established biofilms. Attachment of a hydrophobic palmitic acid tail generates a peptide suited for strong noncovalent surface interactions and allows the generation of stable noncovalent coatings. These adsorbed peptides remain attached to the surface at significant shear stress and also remain active, effectively preventing the biofilm formation over 24 h. Finally, the covalent attachment of the peptides to an acrylate surface was also evaluated and the prepared coatings display a remarkable ability to prevent surface colonization at surface loadings of 55 ng/cm2 over 48 h. The ability to retain the nontoxic antibiofilm activity, documented in solution, in the covalent 2D-format is unprecedented, and this natural peptide motif displays high potential in several material application areas.


Assuntos
Antibacterianos/farmacologia , Bactérias/efeitos dos fármacos , Materiais Biocompatíveis/farmacologia , Venenos de Cnidários/farmacologia , Peptídeos/farmacologia , Animais , Antibacterianos/química , Materiais Biocompatíveis/química , Biofilmes/efeitos dos fármacos , Venenos de Cnidários/química , Teste de Materiais , Testes de Sensibilidade Microbiana , Tamanho da Partícula , Peptídeos/química , Conformação Proteica , Anêmonas-do-Mar/química , Propriedades de Superfície
8.
IEEE/ACM Trans Comput Biol Bioinform ; 18(6): 2816-2822, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33017286

RESUMO

Studying biological systems is a difficult but important task. Traditional methods include laboratory experimentation and computer simulations. However, often researchers need to explore important but potentially rare events that are not easily observed or simulated. We use UPPAAL-SMC, a formal verification tool to support a methodology that allows us to model biological systems, specify events and conditions that we want to analyze, and to explore system executions using controlled simulations. We also describe an efficient way to reproduce laboratory experiments in silico. Unlike traditional simulations, we are able to guide the experiment to explore special events and conditions by expressing these conditions in temporal logic formulas. We have applied this methodology to create a more detailed model of Palytoxin-induced Na +/K + pump channels than was previously possible. Moreover, we have reproduced experimental protocols and their associated electrophysiological recordings, which has not been done in previous works. As a consequence, we have been able to propose a new diprotomeric model for the PTX-pump complex and study its behaviour. The use of our methodology has enabled us to reduce the effort and time to perform this research. It can be used to model and analyze other complex biological systems, potentially increasing the productivity of such studies.


Assuntos
Acrilamidas/farmacologia , Venenos de Cnidários/farmacologia , Biologia Computacional/métodos , Modelos Teóricos , ATPase Trocadora de Sódio-Potássio/efeitos dos fármacos , Processos Estocásticos
9.
Molecules ; 25(24)2020 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-33348592

RESUMO

Actinoporins are the most abundant group of sea anemone cytolytic toxins. Their membranolytic activity is of high interest for the development of novel anticancer drugs. However, to date the activity of actinoporins in malignant cells has been poorly studied. Here, we report on recombinant analog of Hct-S3 (rHct-S3), belonging to the combinatory library of Heteractis crispa actinoporins. rHct-S3 exhibited cytotoxic activity against breast MDA-MB-231 (IC50 = 7.3 µM), colorectal HT-29 (IC50 = 6.8 µM), and melanoma SK-MEL-28 (IC50 = 8.3 µM) cancer cells. The actinoporin effectively prevented epidermal growth factor -induced neoplastic transformation of JB6 Cl41 cells by 34% ± 0.2 and decreased colony formation of HT-29 cells by 47% ± 0.9, MDA-MB-231 cells by 37% ± 1.2, and SK-MEL-28 cells by 34% ± 3.6. Moreover, rHct-S3 decreased proliferation and suppressed migration of colorectal carcinoma cells by 31% ± 5.0 and 99% ± 6.4, respectively. The potent anti-migratory activity was proposed to mediate by decreased matrix metalloproteinases-2 and -9 expression. In addition, rHct-S3 induced programmed cell death by cleavage of caspase-3 and poly (ADP-ribose) polymerase, as well as regulation of Bax and Bcl-2. Our results indicate rHct-S3 to be a promising anticancer drug with a high anti-migratory potential.


Assuntos
Antineoplásicos/farmacologia , Movimento Celular/efeitos dos fármacos , Venenos de Cnidários/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Anêmonas-do-Mar/metabolismo , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Caspase 3/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Transformação Celular Neoplásica/efeitos dos fármacos , Neoplasias Colorretais/patologia , Feminino , Células HT29 , Humanos , Metaloproteinase 2 da Matriz/biossíntese , Metaloproteinase 9 da Matriz/biossíntese , Melanoma/tratamento farmacológico , Melanoma/patologia , Poli(ADP-Ribose) Polimerases/metabolismo
10.
Biomed Pharmacother ; 132: 110807, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33068939

RESUMO

Colorectal cancer (CRC) is a stem cell-based disease. PIK3CA/KRAS-mutant CRC stem cells (CRCSCs) display high self-renewal, metastatic properties, high activity of PI3K and KRAS signaling pathways with chemoresistant phenotypes. Recently, RGD peptide (containing Arg-Gly-Asp motif)-based therapy of solid tumor cells has attracted much attention. However, little is known whether this method can target self-renewal capacity, key effectors of PI3K and KRAS signaling pathways such as metastasis-driver gene CXCR4 and stem cell regulatory genes with caspase-3 reactivation in CRCSCs overexpressing RGD-dependent integrins. The sea anemone Actinia fragacea produces a water-soluble RGD-peptide fragacea toxin C (FraC) suggesting the possible activity of FraC against PIK3CA/KRAS-mutant CRCSCs. Recombinant FraC was expressed via pET-28a(+)-FraC in E. coli and purified through affinity chromatography followed by performing SDS-PAGE and hemolytic activity assay. Next, PIK3CA/KRAS-mutant HCT-116 cells that serve as an attractive model for CRCSCs were treated with FraC. Thereafter, cell numbers, viability, proliferation, LDH activity, cytotoxicity index, CXCR4 and pluripotency network genes expression, self-renewal capacity, caspase-3 activity with apoptosis were evaluated. Caspase-1, -2, -3,…, -9 sequences were analyzed for RGD-binding motifs. FraC sequence and structure were also evaluated by bioinformatics software. FraC altered cellular morphology to round shapes and disrupted cell connections. 48 h post-treatment with 0.056- to 7.2 µM FraC resulted in 12 %-99 % and 8 %-97.6 % decreases in cell numbers and viabilities respectively and increased LDH activity by 0.2 %-66.7 % in a dose-dependent manner. The results of the cytotoxicity index showed that FraC induces significant toxicity on HCT-116 cells compared to PBMCs and Huvec cells. FraC dramatically decreased the expression of CXCR4 and pluripotency network genes Bmi-1, Sox-2, Oct-4 and Nanog followed by remarkable decreases in self-renewal capacity ranged from 91- to 0 colonies per well for 0.056- to 3.6 µM FraC after 2 weeks. Caspase-3 was found to contain an RGD-binding motif and its activity increased with increasing FraC concentrations followed by apoptosis induction. Potential RGD-binding motifs for FraC were also found in caspase-1, -7, -8 and -9. Unique advantages of FraC peptide, such as low molecular weight, water solubility, high sensitivity of CRC stem-like cells with more selective toxicity to this compound, targeting tumor cell membrane and self-renewal capacity along with the modulation of CXCR4 and stem cell regulatory genes as upstream and downstream effectors of undruggable PI3K and KRAS signaling pathways may open up avenues for FraC peptide-based therapy of PIK3CA/KRAS-mutant CRCSCs with lower toxicity on healthy cells.


Assuntos
Venenos de Cnidários/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Oligopeptídeos/farmacologia , Anêmonas-do-Mar/metabolismo , Animais , Antineoplásicos/química , Antineoplásicos/isolamento & purificação , Antineoplásicos/farmacologia , Apoptose/genética , Linhagem Celular Tumoral , Autorrenovação Celular/efeitos dos fármacos , Classe I de Fosfatidilinositol 3-Quinases/genética , Venenos de Cnidários/química , Venenos de Cnidários/isolamento & purificação , Neoplasias Colorretais/genética , Genes Reguladores/genética , Células HCT116 , Humanos , Mutação , Células-Tronco Neoplásicas/citologia , Oligopeptídeos/química , Oligopeptídeos/isolamento & purificação , Proteínas Proto-Oncogênicas p21(ras)/genética , Receptores CXCR4/genética , Transdução de Sinais/efeitos dos fármacos , Solubilidade
11.
Dokl Biochem Biophys ; 493(1): 215-217, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32894469

RESUMO

The ion channel TRPV1, which is one of the most important integrators of pain and inflammatory stimuli, is considered a promising therapeutic target in the treatment of pain conditions. In this work, we performed a comparative study of the analgesic effect in the "hot plate" test of recombinant analogues of Kunitz-type peptides from the sea anemone Heteractis crispa venom: APHC1-modulator of TRPV1 and HCRG21-a full blocker of TRPV1. As a result of biological tests, it was shown that the full blocker HCRG21, despite the higher value of 50% effective concentration of TRPV1 inhibition, had an equal analgesic ability with the APHC1 upon intramuscular administration and retained it for 13 h of observation. The analgesic effect of APHC1 at a dose of 0.1 mg/kg when administered intramuscularly developed very quickly in 5 min but lasted 3 h. The differences in the pharmacodynamic profile of the peptides are in good agreement with different mechanisms of binding to TRPV1.


Assuntos
Analgésicos/farmacologia , Venenos de Cnidários/farmacologia , Dor/tratamento farmacológico , Peptídeos/farmacologia , Canais de Cátion TRPV/antagonistas & inibidores , Sequência de Aminoácidos , Analgésicos/administração & dosagem , Animais , Venenos de Cnidários/administração & dosagem , Modelos Animais de Doenças , Temperatura Alta , Injeções Intramusculares , Camundongos , Camundongos Endogâmicos ICR , Dor/metabolismo , Peptídeos/administração & dosagem , Anêmonas-do-Mar , Homologia de Sequência
12.
Int J Mol Sci ; 21(16)2020 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-32823835

RESUMO

The marine polyether palytoxin (PLTX) is one of the most toxic natural compounds, and is involved in human poisonings after oral, inhalation, skin and/or ocular exposure. Epidemiological and molecular evidence suggest different inter-individual sensitivities to its toxic effects, possibly related to genetic-dependent differences in the expression of Na+/K+-ATPase, its molecular target. To identify Na+/K+-ATPase subunits, isoforms correlated with in vitro PLTX cytotoxic potency, sensitivity parameters (EC50: PLTX concentration reducing cell viability by 50%; Emax: maximum effect induced by the highest toxin concentration; 10-7 M) were assessed in 60 healthy donors' monocytes by the MTT (methylthiazolyl tetrazolium) assay. Sensitivity parameters, not correlated with donors' demographic variables (gender, age and blood group), demonstrated a high inter-individual variability (median EC50 = 2.7 × 10-10 M, interquartile range: 0.4-13.2 × 10-10 M; median Emax = 92.0%, interquartile range: 87.5-94.4%). Spearman's analysis showed significant positive correlations between the ß2-encoding ATP1B2 gene expression and Emax values (rho = 0.30; p = 0.025) and between Emax and the ATP1B2/ATP1B3 expression ratio (rho = 0.38; p = 0.004), as well as a significant negative correlation between Emax and the ATP1B1/ATP1B2 expression ratio (rho = -0.30; p = 0.026). This toxicogenetic study represents the first approach to define genetic risk factors that may influence the onset of adverse effects in human PLTX poisonings, suggesting that individuals with high gene expression pattern of the Na+/K+-ATPase ß2 subunit (alone or as ß2/ß1 and/or ß2/ß3 ratio) could be highly sensitive to PLTX toxic effects.


Assuntos
Acrilamidas/farmacologia , Adenosina Trifosfatases/genética , Proteínas de Transporte de Cátions/genética , Moléculas de Adesão Celular Neuronais/genética , Venenos de Cnidários/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Subunidades Proteicas/genética , Adenosina Trifosfatases/metabolismo , Adulto , Proteínas de Transporte de Cátions/metabolismo , Moléculas de Adesão Celular Neuronais/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Monócitos/efeitos dos fármacos , Monócitos/enzimologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Subunidades Proteicas/metabolismo
13.
Toxins (Basel) ; 12(4)2020 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-32326130

RESUMO

Currently, five peptide modulators of acid-sensing ion channels (ASICs) attributed to structural class 1b of sea anemone toxins have been described. The APETx2 toxin is the first and most potent ASIC3 inhibitor, so its homologs from sea anemones are known as the APETx-like peptides. We have discovered that two APETx-like peptides from the sea anemone Heteractis crispa, Hcr 1b-3 and Hcr 1b-4, demonstrate different effects on rASIC1a and rASIC3 currents. While Hcr 1b-3 inhibits both investigated ASIC subtypes with IC50 4.95 ± 0.19 µM for rASIC1a and 17 ± 5.8 µM for rASIC3, Hcr 1b-4 has been found to be the first potentiator of ASIC3, simultaneously inhibiting rASIC1a at similar concentrations: EC50 1.53 ± 0.07 µM and IC50 1.25 ± 0.04 µM. The closest homologs, APETx2, Hcr 1b-1, and Hcr 1b-2, previously demonstrated the ability to inhibit hASIC3 with IC50 63 nM, 5.5, and 15.9 µM, respectively, while Hcr 1b-2 also inhibited rASIC1a with IC50 4.8 ± 0.3 µM. Computer modeling allowed us to describe the peculiarities of Hcr 1b-2 and Hcr 1b-4 interfaces with the rASIC1a channel and the stabilization of the expanded acidic pocket resulting from peptides binding which traps the rASIC1a channel in the closed state.


Assuntos
Canais Iônicos Sensíveis a Ácido/fisiologia , Venenos de Cnidários/farmacologia , Peptídeos/farmacologia , Anêmonas-do-Mar , Animais , Venenos de Cnidários/química , Modelos Moleculares , Oócitos , Peptídeos/química , Proteínas Recombinantes , Xenopus laevis
14.
Sci Rep ; 9(1): 19307, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31848433

RESUMO

ShK is a 35-residue disulfide-linked polypeptide produced by the sea anemone Stichodactyla helianthus, which blocks the potassium channels Kv1.1 and Kv1.3 with pM affinity. An analogue of ShK has been developed that blocks Kv1.3 > 100 times more potently than Kv1.1, and has completed Phase 1b clinical trials for the treatment of autoimmune diseases such as psoriasis and rheumatoid arthritis. Previous studies have indicated that ShK undergoes a conformational exchange that is critical to its function, but this has proved difficult to characterise. Here, we have used high hydrostatic pressure as a tool to increase the population of the alternative state, which is likely to resemble the active form that binds to the Kv1.3 channel. By following changes in chemical shift with pressure, we have derived the chemical shift values of the low- and high-pressure states, and thus characterised the locations of structural changes. The main difference is in the conformation of the Cys17-Cys32 disulfide, which is likely to affect the positions of the critical Lys22-Tyr23 pair by twisting the 21-24 helix and increasing the solvent exposure of the Lys22 sidechain, as indicated by molecular dynamics simulations.


Assuntos
Venenos de Cnidários/química , Canal de Potássio Kv1.1/antagonistas & inibidores , Canal de Potássio Kv1.3/antagonistas & inibidores , Bloqueadores dos Canais de Potássio/química , Sequência de Aminoácidos/genética , Animais , Doenças Autoimunes/tratamento farmacológico , Venenos de Cnidários/genética , Venenos de Cnidários/farmacologia , Humanos , Canal de Potássio Kv1.1/química , Canal de Potássio Kv1.1/ultraestrutura , Canal de Potássio Kv1.3/química , Canal de Potássio Kv1.3/ultraestrutura , Conformação Molecular , Simulação de Dinâmica Molecular , Peptídeos/química , Peptídeos/genética , Bloqueadores dos Canais de Potássio/farmacologia , Anêmonas-do-Mar/química
15.
Mar Drugs ; 17(10)2019 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-31546678

RESUMO

Sea anemones' venom is rich in peptides acting on different biological targets, mainly on cytoplasmic membranes and ion channels. These animals are also a source of pancreatic α-amylase inhibitors, which have the ability to control the glucose level in the blood and can be used for the treatment of prediabetes and type 2 diabetes mellitus. Recently we have isolated and characterized magnificamide (44 aa, 4770 Da), the major α-amylase inhibitor of the sea anemone Heteractis magnifica mucus, which shares 84% sequence identity with helianthamide from Stichodactyla helianthus. Herein, we report some features in the action of a recombinant analog of magnificamide. The recombinant peptide inhibits porcine pancreatic and human saliva α-amylases with Ki's equal to 0.17 ± 0.06 nM and 7.7 ± 1.5 nM, respectively, and does not show antimicrobial or channel modulating activities. We have concluded that the main function of magnificamide is the inhibition of α-amylases; therefore, its functionally active recombinant analog is a promising agent for further studies as a potential drug candidate for the treatment of the type 2 diabetes mellitus.


Assuntos
Muco/química , Peptídeos/farmacologia , Anêmonas-do-Mar/química , alfa-Amilases/antagonistas & inibidores , beta-Defensinas/farmacologia , Sequência de Aminoácidos , Animais , Glicemia/efeitos dos fármacos , Venenos de Cnidários/farmacologia , Diabetes Mellitus Tipo 2/tratamento farmacológico , Humanos
16.
Neuromolecular Med ; 21(4): 454-466, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31134484

RESUMO

Diabetes is a chronic metabolic disease and cerebral ischemia is a serious complication of diabetes. Anti-diabetic therapy mitigates this complication but increases the risk of exposure to recurrent hypoglycemia (RH). We showed previously that RH exposure increases ischemic brain damage in insulin-treated diabetic (ITD) rats. The present study evaluated the hypothesis that increased intra-ischemic acidosis in RH-exposed ITD rats leads to pronounced post-ischemic hypoperfusion via activation of acid-sensing (proton-gated) ion channels (ASICs). Streptozotocin-diabetic rats treated with insulin were considered ITD rats. ITD rats were exposed to RH for 5 days and were randomized into Psalmotoxin1 (PcTx1, ASIC1a inhibitor), APETx2 (ASIC3 inhibitor), or vehicle groups. Transient global cerebral ischemia was induced overnight after RH. Cerebral blood flow was measured using laser Doppler flowmetry. Ischemic brain injury in hippocampus was evaluated using histopathology. Post-ischemic hypoperfusion in RH-exposed rats was of greater extent than that in control rats. Inhibition of ASICs prevented RH-induced increase in the extent of post-ischemic hypoperfusion and ischemic brain injury. Since ASIC activation-induced store-operated calcium entry (SOCE) plays a role in vascular tone, next we tested if acidosis activates SOCE via activating ASICs in vascular smooth muscle cells (VSMCs). We observed that SOCE in VSMCs at lower pH is ASIC3 dependent. The results show the role of ASIC in post-ischemic hypoperfusion and increased ischemic damage in RH-exposed ITD rats. Understanding the pathways mediating exacerbated ischemic brain injury in RH-exposed ITD rats may help lower diabetic aggravation of ischemic brain damage.


Assuntos
Bloqueadores do Canal Iônico Sensível a Ácido/uso terapêutico , Canais Iônicos Sensíveis a Ácido/fisiologia , Acidose/tratamento farmacológico , Dano Encefálico Crônico/prevenção & controle , Isquemia Encefálica/complicações , Estenose das Carótidas/complicações , Venenos de Cnidários/uso terapêutico , Diabetes Mellitus Experimental/complicações , Hipoglicemia/complicações , Hipoglicemiantes/toxicidade , Insulina/toxicidade , Peptídeos/uso terapêutico , Venenos de Aranha/uso terapêutico , Bloqueadores do Canal Iônico Sensível a Ácido/farmacologia , Canais Iônicos Sensíveis a Ácido/efeitos dos fármacos , Acidose/etiologia , Animais , Dano Encefálico Crônico/etiologia , Isquemia Encefálica/fisiopatologia , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Circulação Cerebrovascular , Venenos de Cnidários/farmacologia , Diabetes Mellitus Experimental/tratamento farmacológico , Hipoglicemia/sangue , Hipoglicemia/induzido quimicamente , Hipoglicemiantes/uso terapêutico , Insulina/uso terapêutico , Fluxometria por Laser-Doppler , Masculino , Peptídeos/farmacologia , Distribuição Aleatória , Ratos , Ratos Wistar , Recidiva , Venenos de Aranha/farmacologia
17.
Pain Med ; 20(10): 1963-1970, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30908578

RESUMO

BACKGROUND: Low-level laser therapy (LLLT) is widely used in pain control in the field of physical medicine and rehabilitation and is effective for fibromyalgia pain. However, its analgesic mechanism remains unknown. A possible mechanism for the effect of LLLT on fibromyalgia pain is via the antinociceptive signaling of substance P in muscle nociceptors, although the neuropeptide has been known as a neurotransmitter to facilitate pain signals in the spinal cord. OBJECTIVE: To establish an animal model of LLLT in chronic muscle pain and to determine the role of substance P in LLLT analgesia. METHODS: We employed the acid-induced chronic muscle pain model, a fibromyalgia model proposed and developed by Sluka et al., and determined the optimal LLLT dosage. RESULTS: LLLT with 685 nm at 8 J/cm2 was effective to reduce mechanical hyperalgesia in the chronic muscle pain model. The analgesic effect was abolished by pretreatment of NK1 receptor antagonist RP-67580. Likewise, LLLT showed no analgesic effect on Tac1-/- mice, in which the gene encoding substance P was deleted. Besides, pretreatment with the TRPV1 receptor antagonist capsazepine, but not the ASIC3 antagonist APETx2, blocked the LLLT analgesic effect. CONCLUSIONS: LLLT analgesia is mediated by the antinociceptive signaling of intramuscular substance P and is associated with TRPV1 activation in a mouse model of fibromyalgia or chronic muscle pain. The study results could provide new insight regarding the effect of LLLT in other types of chronic pain.


Assuntos
Terapia a Laser , Dor Musculoesquelética/metabolismo , Dor Musculoesquelética/terapia , Substância P/fisiologia , Ácidos , Animais , Capsaicina/análogos & derivados , Capsaicina/farmacologia , Dor Crônica/metabolismo , Dor Crônica/terapia , Venenos de Cnidários/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Fibromialgia/induzido quimicamente , Fibromialgia/psicologia , Fibromialgia/terapia , Terapia com Luz de Baixa Intensidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dor Musculoesquelética/induzido quimicamente , Precursores de Proteínas/genética , Transdução de Sinais , Canais de Cátion TRPV/efeitos dos fármacos , Taquicininas/genética
18.
Biochimie ; 156: 206-223, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30036605

RESUMO

Actinoporins constitute a unique class of pore-forming toxins found in sea anemones that being secreted as soluble monomers are able to bind and permeabilize membranes leading to cell death. The interest in these proteins has risen due to their high cytotoxicity that can be properly used to design immunotoxins against tumor cells and antigen-releasing systems to cell cytosol. In this work we describe a novel actinoporin produced by Anthopleura nigrescens, an anemone found in the Central American Pacific Ocean. Here we report the amino acid sequence of an actinoporin as deduced from cDNA obtained from total body RNA. The synthetic DNA sequence encoding for one cytolysin variant was expressed in BL21 Star (DE3) Escherichia coli and the protein purified by chromatography on CM Sephadex C-25 with more than 97% homogeneity as verified by MS-MS and HPLC analyses. This actinoporin comprises 179 amino acid residues, consistent with its observed isotope-averaged molecular mass of 19 661 Da. The toxin lacks Cys and readily permeabilizes erythrocytes, as well as L1210 cells. CD spectroscopy revealed that its secondary structure is dominated by beta structure (58.5%) with 5.5% of α-helix, and 35% of random structure. Moreover, binding experiments to lipidic monolayers and to liposomes, as well as permeabilization studies in vesicles, revealed that the affinity of this toxin for sphingomyelin-containing membranes is quite similar to sticholysin II (StII). Comparison by spectroscopic techniques and modeling the three-dimensional structure of nigrelysin (Ng) showed a high homology with StII but several differences were also detectable. Taken together, these results reinforce the notion that Ng is a novel member of the actinoporin pore-forming toxin (PFT) family with a HA as high as that of StII, the most potent actinoporin so far described, but with peculiar structural characteristics contributing to expand the understanding of the structure-function relationship in this protein family.


Assuntos
Permeabilidade da Membrana Celular/efeitos dos fármacos , Venenos de Cnidários , Membrana Eritrocítica , Membranas Artificiais , Anemone/química , Anemone/genética , Clonagem Molecular , Venenos de Cnidários/biossíntese , Venenos de Cnidários/química , Venenos de Cnidários/genética , Venenos de Cnidários/farmacologia , Membrana Eritrocítica/química , Membrana Eritrocítica/metabolismo , Humanos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia
19.
ACS Chem Biol ; 13(11): 3153-3160, 2018 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-30278129

RESUMO

Immunotoxins are proteins containing a cell-targeting element linked to a toxin that are under investigation for next-generation cancer treatment. However, these agents are difficult to synthesize, chemically heterogeneous, expensive, and show toxicity toward healthy cells. In this work, we describe the synthesis and characterization of a new type of immunotoxin that showed exquisite selectivity toward targeted cells. In our construct, targeting molecules were covalently attached or genetically fused to oligomeric pore-forming toxins. The activity of the immunotoxin was then caged by fusing a soluble protein to the transmembrane domain and activated via cleavage with furin, which is a protease that is overexpressed in many cancer cells. During the several coupling steps, directed evolution allowed the efficient synthesis of the molecules in E. coli cells, as well as selection for further specificity toward targeted cells. The final construct showed no off-target activity, while acquiring an additional degree of specificity toward the targeted cells upon activation. The pore-forming toxins described here do not require internalization to operate, while the many protomeric subunits can be individually modified to refine target specificity.


Assuntos
Venenos de Cnidários/farmacologia , Imunotoxinas/farmacologia , Proteínas Citotóxicas Formadoras de Poros/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Tetra-Hidrofolato Desidrogenase/farmacologia , Animais , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Venenos de Cnidários/genética , Evolução Molecular Direcionada/métodos , Desenho de Fármacos , Escherichia coli/genética , Escherichia coli/metabolismo , Ácido Fólico/química , Furina/metabolismo , Humanos , Imunotoxinas/química , Imunotoxinas/genética , Imunotoxinas/metabolismo , Mutagênese , Proteínas Citotóxicas Formadoras de Poros/genética , Proteólise , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Salmonella typhi/química , Anêmonas-do-Mar/química , Tetra-Hidrofolato Desidrogenase/genética , Tetra-Hidrofolato Desidrogenase/metabolismo
20.
Mar Drugs ; 16(10)2018 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-30275388

RESUMO

Voltage-gated potassium (KV) channels regulate diverse physiological processes and are an important target for developing novel therapeutic approaches. Sea anemone (Cnidaria, Anthozoa) venoms comprise a highly complex mixture of peptide toxins with diverse and selective pharmacology on KV channels. From the nematocysts of the sea anemone Actinia bermudensis, a peptide that we named AbeTx1 was purified and functionally characterized on 12 different subtypes of KV channels (KV1.1⁻KV1.6; KV2.1; KV3.1; KV4.2; KV4.3; KV11.1; and, Shaker IR), and three voltage-gated sodium channel isoforms (NaV1.2, NaV1.4, and BgNaV). AbeTx1 was selective for Shaker-related K⁺ channels and is capable of inhibiting K⁺ currents, not only by blocking the K⁺ current of KV1.2 subtype, but by altering the energetics of activation of KV1.1 and KV1.6. Moreover, experiments using six synthetic alanine point-mutated analogs further showed that a ring of basic amino acids acts as a multipoint interaction for the binding of the toxin to the channel. The AbeTx1 primary sequence is composed of 17 amino acids with a high proportion of lysines and arginines, including two disulfide bridges (Cys1⁻Cys4 and Cys2⁻Cys3), and it is devoid of aromatic or aliphatic amino acids. Secondary structure analysis reveals that AbeTx1 has a highly flexible, random-coil-like conformation, but with a tendency of structuring in the beta sheet. Its overall structure is similar to open-ended cyclic peptides found on the scorpion κ-KTx toxins family, cone snail venoms, and antimicrobial peptides.


Assuntos
Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Anêmonas-do-Mar/química , Anêmonas-do-Mar/metabolismo , Toxinas Biológicas/química , Toxinas Biológicas/farmacologia , Sequência de Aminoácidos , Aminoácidos/metabolismo , Animais , Venenos de Cnidários/química , Venenos de Cnidários/farmacologia , Peptídeos/química , Peptídeos/farmacologia , Potássio/metabolismo , Bloqueadores dos Canais de Potássio/química , Bloqueadores dos Canais de Potássio/farmacologia , Alinhamento de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA