Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(5)2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35074873

RESUMEN

The King Baboon spider, Pelinobius muticus, is a burrowing African tarantula. Its impressive size and appealing coloration are tempered by reports describing severe localized pain, swelling, itchiness, and muscle cramping after accidental envenomation. Hyperalgesia is the most prominent symptom after bites from P. muticus, but the molecular basis by which the venom induces pain is unknown. Proteotranscriptomic analysis of P. muticus venom uncovered a cysteine-rich peptide, δ/κ-theraphotoxin-Pm1a (δ/κ-TRTX-Pm1a), that elicited nocifensive behavior when injected into mice. In small dorsal root ganglion neurons, synthetic δ/κ-TRTX-Pm1a (sPm1a) induced hyperexcitability by enhancing tetrodotoxin-resistant sodium currents, impairing repolarization and lowering the threshold of action potential firing, consistent with the severe pain associated with envenomation. The molecular mechanism of nociceptor sensitization by sPm1a involves multimodal actions over several ion channel targets, including NaV1.8, KV2.1, and tetrodotoxin-sensitive NaV channels. The promiscuous targeting of peptides like δ/κ-TRTX-Pm1a may be an evolutionary adaptation in pain-inducing defensive venoms.


Asunto(s)
Nociceptores/efectos de los fármacos , Papio/metabolismo , Péptidos/farmacología , Venenos de Araña/farmacología , Arañas/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Ganglios Espinales/efectos de los fármacos , Hiperalgesia/tratamiento farmacológico , Canales Iónicos/metabolismo , Ratones , Dolor/tratamiento farmacológico , Tetrodotoxina/farmacología
2.
Proc Natl Acad Sci U S A ; 118(18)2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-33893140

RESUMEN

Venoms have evolved independently several times in Lepidoptera. Limacodidae is a family with worldwide distribution, many of which are venomous in the larval stage, but the composition and mode of action of their venom is unknown. Here, we use imaging technologies, transcriptomics, proteomics, and functional assays to provide a holistic picture of the venom system of a limacodid caterpillar, Doratifera vulnerans Contrary to dogma that defensive venoms are simple in composition, D. vulnerans produces a complex venom containing 151 proteinaceous toxins spanning 59 families, most of which are peptides <10 kDa. Three of the most abundant families of venom peptides (vulnericins) are 1) analogs of the adipokinetic hormone/corazonin-related neuropeptide, some of which are picomolar agonists of the endogenous insect receptor; 2) linear cationic peptides derived from cecropin, an insect innate immune peptide that kills bacteria and parasites by disrupting cell membranes; and 3) disulfide-rich knottins similar to those that dominate spider venoms. Using venom fractionation and a suite of synthetic venom peptides, we demonstrate that the cecropin-like peptides are responsible for the dominant pain effect observed in mammalian in vitro and in vivo nociception assays and therefore are likely to cause pain after natural envenomations by D. vulnerans Our data reveal convergent molecular evolution between limacodids, hymenopterans, and arachnids and demonstrate that lepidopteran venoms are an untapped source of novel bioactive peptides.


Asunto(s)
Venenos de Artrópodos/química , Proteínas de Insectos/química , Lepidópteros/química , Neuropéptidos/química , Dolor/genética , Animales , Venenos de Artrópodos/genética , Evolución Molecular , Proteínas de Insectos/genética , Mariposas Nocturnas/química , Neuropéptidos/genética , Péptidos/química , Péptidos/genética , Proteómica , Venenos de Araña/química , Venenos de Araña/genética , Transcriptoma/genética
3.
Int J Mol Sci ; 25(9)2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38731963

RESUMEN

Venom peptides have evolved to target a wide range of membrane proteins through diverse mechanisms of action and structures, providing promising therapeutic leads for diseases, including pain, epilepsy, and cancer, as well as unique probes of ion channel structure-function. In this work, a high-throughput FLIPR window current screening assay on T-type CaV3.2 guided the isolation of a novel peptide named ω-Buthitoxin-Hf1a from scorpion Hottentotta franzwerneri crude venom. At only 10 amino acid residues with one disulfide bond, it is not only the smallest venom peptide known to target T-type CaVs but also the smallest structured scorpion venom peptide yet discovered. Synthetic Hf1a peptides were prepared with C-terminal amidation (Hf1a-NH2) or a free C-terminus (Hf1a-OH). Electrophysiological characterization revealed Hf1a-NH2 to be a concentration-dependent partial inhibitor of CaV3.2 (IC50 = 1.18 µM) and CaV3.3 (IC50 = 0.49 µM) depolarized currents but was ineffective at CaV3.1. Hf1a-OH did not show activity against any of the three T-type subtypes. Additionally, neither form showed activity against N-type CaV2.2 or L-type calcium channels. The three-dimensional structure of Hf1a-NH2 was determined using NMR spectroscopy and used in docking studies to predict its binding site at CaV3.2 and CaV3.3. As both CaV3.2 and CaV3.3 have been implicated in peripheral pain signaling, the analgesic potential of Hf1a-NH2 was explored in vivo in a mouse model of incision-induced acute post-surgical pain. Consistent with this role, Hf1a-NH2 produced antiallodynia in both mechanical and thermal pain.


Asunto(s)
Canales de Calcio Tipo T , Modelos Animales de Enfermedad , Hiperalgesia , Dolor Postoperatorio , Venenos de Escorpión , Animales , Canales de Calcio Tipo T/metabolismo , Canales de Calcio Tipo T/química , Ratones , Venenos de Escorpión/química , Venenos de Escorpión/farmacología , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , Dolor Postoperatorio/tratamiento farmacológico , Dolor Postoperatorio/metabolismo , Calcio/metabolismo , Masculino , Humanos , Bloqueadores de los Canales de Calcio/farmacología , Bloqueadores de los Canales de Calcio/química
4.
Nature ; 534(7608): 494-9, 2016 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-27281198

RESUMEN

Voltage-gated sodium (Nav) channels initiate action potentials in most neurons, including primary afferent nerve fibres of the pain pathway. Local anaesthetics block pain through non-specific actions at all Nav channels, but the discovery of selective modulators would facilitate the analysis of individual subtypes of these channels and their contributions to chemical, mechanical, or thermal pain. Here we identify and characterize spider (Heteroscodra maculata) toxins that selectively activate the Nav1.1 subtype, the role of which in nociception and pain has not been elucidated. We use these probes to show that Nav1.1-expressing fibres are modality-specific nociceptors: their activation elicits robust pain behaviours without neurogenic inflammation and produces profound hypersensitivity to mechanical, but not thermal, stimuli. In the gut, high-threshold mechanosensitive fibres also express Nav1.1 and show enhanced toxin sensitivity in a mouse model of irritable bowel syndrome. Together, these findings establish an unexpected role for Nav1.1 channels in regulating the excitability of sensory nerve fibres that mediate mechanical pain.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Nocicepción/efectos de los fármacos , Nociceptores/efectos de los fármacos , Nociceptores/metabolismo , Venenos de Araña/farmacología , Estrés Mecánico , Animales , Modelos Animales de Enfermedad , Femenino , Ganglios Sensoriales/citología , Hiperalgesia/inducido químicamente , Hiperalgesia/metabolismo , Síndrome del Colon Irritable/metabolismo , Masculino , Vaina de Mielina/metabolismo , Canal de Sodio Activado por Voltaje NAV1.1/química , Fibras Nerviosas/efectos de los fármacos , Fibras Nerviosas/metabolismo , Oocitos/metabolismo , Dolor/inducido químicamente , Dolor/metabolismo , Estructura Terciaria de Proteína , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo , Arañas/química , Especificidad por Sustrato/efectos de los fármacos , Temperatura
5.
Proc Natl Acad Sci U S A ; 116(44): 22353-22358, 2019 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-31611414

RESUMEN

An Australian estuarine isolate of Penicillium sp. MST-MF667 yielded 3 tetrapeptides named the bilaids with an unusual alternating LDLD chirality. Given their resemblance to known short peptide opioid agonists, we elucidated that they were weak (Ki low micromolar) µ-opioid agonists, which led to the design of bilorphin, a potent and selective µ-opioid receptor (MOPr) agonist (Ki 1.1 nM). In sharp contrast to all-natural product opioid peptides that efficaciously recruit ß-arrestin, bilorphin is G protein biased, weakly phosphorylating the MOPr and marginally recruiting ß-arrestin, with no receptor internalization. Importantly, bilorphin exhibits a similar G protein bias to oliceridine, a small nonpeptide with improved overdose safety. Molecular dynamics simulations of bilorphin and the strongly arrestin-biased endomorphin-2 with the MOPr indicate distinct receptor interactions and receptor conformations that could underlie their large differences in bias. Whereas bilorphin is systemically inactive, a glycosylated analog, bilactorphin, is orally active with similar in vivo potency to morphine. Bilorphin is both a unique molecular tool that enhances understanding of MOPr biased signaling and a promising lead in the development of next generation analgesics.


Asunto(s)
Analgésicos Opioides/farmacología , Proteínas Fúngicas/farmacología , Oligopéptidos/farmacología , Penicillium/química , Receptores Opioides mu/agonistas , Analgésicos Opioides/química , Animales , Sitios de Unión , Línea Celular Tumoral , Proteínas Fúngicas/química , Células HEK293 , Humanos , Ratones , Simulación del Acoplamiento Molecular , Oligopéptidos/química , Unión Proteica , Receptores Opioides mu/química , Receptores Opioides mu/metabolismo
6.
Cell Mol Life Sci ; 75(24): 4511-4524, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30109357

RESUMEN

Sea anemone venoms have long been recognized as a rich source of peptides with interesting pharmacological and structural properties, but they still contain many uncharacterized bioactive compounds. Here we report the discovery, three-dimensional structure, activity, tissue localization, and putative function of a novel sea anemone peptide toxin that constitutes a new, sixth type of voltage-gated potassium channel (KV) toxin from sea anemones. Comprised of just 17 residues, κ-actitoxin-Ate1a (Ate1a) is the shortest sea anemone toxin reported to date, and it adopts a novel three-dimensional structure that we have named the Proline-Hinged Asymmetric ß-hairpin (PHAB) fold. Mass spectrometry imaging and bioassays suggest that Ate1a serves a primarily predatory function by immobilising prey, and we show this is achieved through inhibition of Shaker-type KV channels. Ate1a is encoded as a multi-domain precursor protein that yields multiple identical mature peptides, which likely evolved by multiple domain duplication events in an actinioidean ancestor. Despite this ancient evolutionary history, the PHAB-encoding gene family exhibits remarkable sequence conservation in the mature peptide domains. We demonstrate that this conservation is likely due to intra-gene concerted evolution, which has to our knowledge not previously been reported for toxin genes. We propose that the concerted evolution of toxin domains provides a hitherto unrecognised way to circumvent the effects of the costly evolutionary arms race considered to drive toxin gene evolution by ensuring efficient secretion of ecologically important predatory toxins.


Asunto(s)
Venenos de Cnidarios/química , Péptidos/química , Canales de Potasio con Entrada de Voltaje/química , Anémonas de Mar/química , Secuencia de Aminoácidos , Animales , Venenos de Cnidarios/genética , Venenos de Cnidarios/metabolismo , Evolución Molecular , Modelos Moleculares , Péptidos/genética , Péptidos/metabolismo , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/metabolismo , Conformación Proteica , Pliegue de Proteína , Anémonas de Mar/genética , Anémonas de Mar/metabolismo , Transcriptoma
7.
J Biol Chem ; 291(22): 11829-42, 2016 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-27026701

RESUMEN

The µO-conotoxins MrVIA, MrVIB, and MfVIA inhibit the voltage-gated sodium channel NaV1.8, a well described target for the treatment of pain; however, little is known about the residues or structural elements that define this activity. In this study, we determined the three-dimensional structure of MfVIA, examined its membrane binding properties, performed alanine-scanning mutagenesis, and identified residues important for its activity at human NaV1.8. A second round of mutations resulted in (E5K,E8K)MfVIA, a double mutant with greater positive surface charge and greater affinity for lipid membranes compared with MfVIA. This analogue had increased potency at NaV1.8 and was analgesic in the mouse formalin assay.


Asunto(s)
Analgésicos/farmacología , Membrana Celular/metabolismo , Conotoxinas/farmacología , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Dolor/prevención & control , Secuencia de Aminoácidos , Animales , Conducta Animal/efectos de los fármacos , Cristalografía por Rayos X , Electrofisiología , Células HEK293 , Humanos , Liposomas , Espectroscopía de Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Canal de Sodio Activado por Voltaje NAV1.8/química , Canal de Sodio Activado por Voltaje NAV1.8/genética , Dolor/inducido químicamente , Conformación Proteica , Homología de Secuencia de Aminoácido
8.
Angew Chem Int Ed Engl ; 56(47): 14973-14976, 2017 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-28984021

RESUMEN

Conotoxins are a large family of disulfide-rich peptides that contain unique cysteine frameworks that target a broad range of ion channels and receptors. We recently discovered the 33-residue conotoxin Φ-MiXXVIIA from Conus miles with a novel cysteine framework comprising three consecutive cysteine residues and four disulfide bonds. Regioselective chemical synthesis helped decipher the disulfide bond connectivity and the structure of Φ-MiXXVIIA was determined by NMR spectroscopy. The 3D structure displays a unique topology containing two ß-hairpins that resemble the N-terminal domain of granulin. Similar to granulin, Φ-MiXXVIIA promotes cell proliferation (EC50 17.85 µm) while inhibiting apoptosis (EC50 2.2 µm). Additional framework XXVII sequences were discovered with homologous signal peptides that define the new conotoxin superfamily G2. The novel structure and biological activity of Φ-MiXXVIIA expands the repertoire of disulfide-rich conotoxins that recognize mammalian receptors.


Asunto(s)
Apoptosis/efectos de los fármacos , Conotoxinas/farmacología , Cisteína/química , Granulinas/farmacología , Imitación Molecular , Secuencia de Aminoácidos , Proliferación Celular/efectos de los fármacos , Conotoxinas/química , Disulfuros/química , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Conformación Proteica , Homología de Secuencia de Aminoácido
9.
Angew Chem Int Ed Engl ; 56(29): 8495-8499, 2017 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-28513074

RESUMEN

Δ-Myrtoxin-Mp1a (Mp1a), a 49-residue heterodimeric peptide from the venom of Myrmecia pilosula, comprises a 26-mer A chain and a 23-mer B chain connected by two disulfide bonds in an antiparallel arrangement. Combination of the individual synthetic chains through aerial oxidation remarkably resulted in the self-assembly of Mp1a as a homogenous product without the need for directed disulfide-bond formation. NMR analysis revealed a well-defined, unique structure containing an antiparallel α-helix pair. Dual polarization interferometry (DPI) analysis showed strong interaction with supported lipid bilayers and insertion within the bilayers. Mp1a caused non-specific Ca2+ influx in SH-SY5Y cells with a half maximal effective concentration (EC50 ) of 4.3 µm. Mp1a also displayed broad-spectrum antimicrobial activity, with the highest potency against Gram-negative Acinetobacter baumannii (MIC 25 nm). Intraplantar injection (10 µm) in mice elicited spontaneous pain and mechanical allodynia. Single- and two-chain mimetics of Mp1a revealed functional selectivity.


Asunto(s)
Acinetobacter baumannii/efectos de los fármacos , Antibacterianos/farmacología , Hiperalgesia/tratamiento farmacológico , Dolor/tratamiento farmacológico , Péptidos/farmacología , Ponzoñas/química , Animales , Antibacterianos/administración & dosificación , Antibacterianos/química , Hormigas , Calcio/metabolismo , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Humanos , Ratones , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Péptidos/administración & dosificación , Péptidos/química
10.
Mol Pharmacol ; 88(2): 291-303, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25979003

RESUMEN

Spider venoms are a rich source of ion channel modulators with therapeutic potential. Given the analgesic potential of subtype-selective inhibitors of voltage-gated sodium (NaV) channels, we screened spider venoms for inhibitors of human NaV1.7 (hNaV1.7) using a high-throughput fluorescent assay. Here, we describe the discovery of a novel NaV1.7 inhibitor, µ-TRTX-Tp1a (Tp1a), isolated from the venom of the Peruvian green-velvet tarantula Thrixopelma pruriens. Recombinant and synthetic forms of this 33-residue peptide preferentially inhibited hNaV1.7 > hNaV1.6 > hNaV1.2 > hNaV1.1 > hNaV1.3 channels in fluorescent assays. NaV1.7 inhibition was diminished (IC50 11.5 nM) and the association rate decreased for the C-terminal acid form of Tp1a compared with the native amidated form (IC50 2.1 nM), suggesting that the peptide C terminus contributes to its interaction with hNaV1.7. Tp1a had no effect on human voltage-gated calcium channels or nicotinic acetylcholine receptors at 5 µM. Unlike most spider toxins that modulate NaV channels, Tp1a inhibited hNaV1.7 without significantly altering the voltage dependence of activation or inactivation. Tp1a proved to be analgesic by reversing spontaneous pain induced in mice by intraplantar injection in OD1, a scorpion toxin that potentiates hNaV1.7. The structure of Tp1a as determined using NMR spectroscopy revealed a classic inhibitor cystine knot (ICK) motif. The molecular surface of Tp1a presents a hydrophobic patch surrounded by positively charged residues, with subtle differences from other ICK spider toxins that might contribute to its different pharmacological profile. Tp1a may help guide the development of more selective and potent hNaV1.7 inhibitors for treatment of chronic pain.


Asunto(s)
Analgésicos/farmacología , Dolor/tratamiento farmacológico , Venenos de Araña/farmacología , Arañas/metabolismo , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología , Analgésicos/química , Analgésicos/aislamiento & purificación , Animales , Células CHO , Línea Celular Tumoral , Cricetulus , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Masculino , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Dolor/inducido químicamente , Venenos de Escorpión , Venenos de Araña/química , Venenos de Araña/aislamiento & purificación , Arañas/clasificación , Bloqueadores del Canal de Sodio Activado por Voltaje/química , Bloqueadores del Canal de Sodio Activado por Voltaje/aislamiento & purificación
11.
Angew Chem Int Ed Engl ; 53(11): 2931-4, 2014 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-24604812

RESUMEN

A safety-catch cysteine protecting group, S-4,4'-dimethylsulfinylbenzhydryl (Msbh), was designed and developed to expand the capabilities of synthetic strategies for the regioselective formation of disulfide bonds in cysteine-rich peptides. The directed regioselective synthesis of human hepcidin, which contains four disulfide bonds, was undertaken and led to a high-resolution NMR structure under more physiologically relevant conditions than previously. Conversely, hepcidin synthesized with the formerly assigned vicinal disulfide-bond connectivity displayed significant conformational heterogeneity under similar conditions. The two synthetic forms of human hepcidin induced ferroportin internalization with apparent EC50 values of 2.0 (native fold, 1) and 4.4 nM (non-native fold, 2), with 2 undergoing isomerization to 1 in the presence of ferroportin expressing cells.


Asunto(s)
Compuestos de Bencidrilo/química , Cisteína/química , Disulfuros/química , Hepcidinas/síntesis química , Secuencia de Aminoácidos , Hepcidinas/química , Humanos , Datos de Secuencia Molecular , Pliegue de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Estereoisomerismo
12.
Org Biomol Chem ; 10(30): 5791-4, 2012 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-22581211

RESUMEN

A dual-pharmacophoric peptide was engineered by grafting the integrin binding RGD motif between the C- and N-termini of a disulfide-rich noradrenaline transporter inhibiting χ-conotoxin resulting in a stable backbone cyclized peptide. The construct maintained two independent biological activities and showed increased plasma stability with no adverse effects observed following administration to rats, highlighting the potential value of pharmacophore grafting into constrained peptide scaffolds.


Asunto(s)
Transporte Biológico/efectos de los fármacos , Conotoxinas/metabolismo , Conotoxinas/farmacología , Integrinas/metabolismo , Norepinefrina/metabolismo , Ingeniería de Proteínas/métodos , Animales , Conotoxinas/química , Conotoxinas/farmacocinética , Humanos , Integrinas/antagonistas & inhibidores , Modelos Moleculares , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/antagonistas & inhibidores , Oligopéptidos , Péptidos Cíclicos/metabolismo , Estabilidad Proteica , Ratas
13.
Biomedicines ; 10(5)2022 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-35625803

RESUMEN

Inhibition of T-type calcium channels (CaV3) prevents development of diseases related to cardiovascular and nerve systems. Further, knockout animal studies have revealed that some diseases are mediated by specific subtypes of CaV3. However, subtype-specific CaV3 inhibitors for therapeutic purposes or for studying the physiological roles of CaV3 subtypes are missing. To bridge this gap, we employed our spider venom library and uncovered that Avicularia spec. ("Amazonas Purple", Peru) tarantula venom inhibited specific T-type CaV channel subtypes. By using chromatographic and mass-spectrometric techniques, we isolated and sequenced the active toxin ω-Avsp1a, a C-terminally amidated 36 residue peptide with a molecular weight of 4224.91 Da, which comprised the major peak in the venom. Both native (4.1 µM) and synthetic ω-Avsp1a (10 µM) inhibited 90% of CaV3.1 and CaV3.3, but only 25% of CaV3.2 currents. In order to investigate the toxin binding site, we generated a range of chimeric channels from the less sensitive CaV3.2 and more sensitive CaV3.3. Our results suggest that domain-1 of CaV3.3 is important for the inhibitory effect of ω-Avsp1a on T-type calcium channels. Further studies revealed that a leucine of T-type calcium channels is crucial for the inhibitory effect of ω-Avsp1a.

14.
ACS Chem Neurosci ; 13(8): 1245-1250, 2022 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-35357806

RESUMEN

α-Conotoxins that target muscle nicotinic acetylcholine receptors (nAChRs) commonly fall into two structural classes, frameworks I and II containing two and three disulfide bonds, respectively. Conotoxin SII is the sole member of the cysteine-rich framework II with ill-defined interactions at the nAChRs. Following directed synthesis of α-SII, NMR analysis revealed a well-defined structure containing a 310-helix frequently employed by framework I α-conotoxins; α-SII acted at the muscle nAChR with half-maximal inhibitory concentrations (IC50) of 120 nM (adult) and 370 nM (fetal) though weakly at neuronal nAChRs. Truncation of α-SII to a two disulfide bond amidated peptide with framework I disulfide connectivity led to similar activity. Surprisingly, the more constrained α-SII was less stable under mild reducing conditions and displayed a unique docking mode at the nAChR.


Asunto(s)
Conotoxinas , Receptores Nicotínicos , Secuencia de Aminoácidos , Conotoxinas/farmacología , Cisteína , Disulfuros , Músculos/metabolismo , Antagonistas Nicotínicos/farmacología , Receptores Nicotínicos/metabolismo
15.
J Am Chem Soc ; 133(40): 15866-9, 2011 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-21899353

RESUMEN

The two disulfide bonds of α-conotoxin ImI, a peptide antagonist of the α7 nicotinic acetylcholine receptor (nAChR), were systematically replaced with isosteric redox-stable cystathionine thioethers. Regioselective thioether formation was accomplished on solid support through substitution of a γ-chlorohomoalanine by an intramolecular cysteine thiol to produce hybrid thioether/disulfide analogues (2 and 3) as well as a dual cystathionine analogue (4) that were found to be structurally homologous to α-conotoxin ImI by (1)H NMR. The antagonistic activity at the α7 nAChR of cystathionine analogue 3 (pIC(50) = 6.41 ± 0.09) was identical to that of α-conotoxin ImI (1, pIC(50) = 6.41 ± 0.09), whereas those of 2 (pIC(50) = 5.96 ± 0.09) and 4 (pIC(50) = 5.89 ± 0.09) showed a modest decrease. The effect of oxidation of the thioethers to sulfoxides was also investigated, with significant changes in the biological activities observed ranging from a >30-fold reduction (2S═O) to a 3-fold increase (3S═O(B)) in potencies.


Asunto(s)
Conotoxinas/química , Cistationina/química , Antagonistas Nicotínicos/química , Secuencia de Aminoácidos , Animales , Caracol Conus/química , Modelos Moleculares , Resonancia Magnética Nuclear Biomolecular , Conformación Proteica , Receptores Nicotínicos/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7
16.
Chem Sci ; 12(11): 4057-4062, 2021 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-34163676

RESUMEN

Vasopressin (VP) and oxytocin (OT) are cyclic neuropeptides that regulate fundamental physiological functions via four G protein-coupled receptors, V1aR, V1bR, V2R, and OTR. Ligand development remains challenging for these receptors due to complex structure-activity relationships. Here, we investigated dimerization as a strategy for developing ligands with novel pharmacology. We regioselectively synthesised and systematically studied parallel, antiparallel and N- to C-terminal cyclized homo- and heterodimer constructs of VP, OT and dVDAVP (1-deamino-4-valine-8-d-arginine-VP). All disulfide-linked dimers, except for the head-to-tail cyclized constructs, retained nanomolar potency despite the structural implications of dimerization. Our results support a single chain interaction for receptor activation. Dimer orientation had little impact on activity, except for the dVDAVP homodimers, where an antagonist to agonist switch was observed at the V1aR. This study provides novel insights into the structural requirements of VP/OT receptor activation and spotlights dimerization as a strategy to modulate pharmacology, a concept also frequently observed in nature.

17.
Front Pharmacol ; 12: 789570, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35095499

RESUMEN

Venoms from cone snails and arachnids are a rich source of peptide modulators of voltage-gated sodium (NaV) channels, however relatively few venom-derived peptides with activity at the mammalian NaV1.8 subtype have been isolated. Here, we describe the discovery and functional characterisation of ß-theraphotoxin-Eo1a, a peptide from the venom of the Tanzanian black and olive baboon tarantula Encyocratella olivacea that modulates NaV1.8. Eo1a is a 37-residue peptide that increases NaV1.8 peak current (EC50 894 ± 146 nM) and causes a large hyperpolarising shift in both the voltage-dependence of activation (ΔV50-20.5 ± 1.2 mV) and steady-state fast inactivation (ΔV50-15.5 ± 1.8 mV). At a concentration of 10 µM, Eo1a has varying effects on the peak current and channel gating of NaV1.1-NaV1.7, although its activity is most pronounced at NaV1.8. Investigations into the binding site of Eo1a using NaV1.7/NaV1.8 chimeras revealed a critical contribution of the DII S3-S4 extracellular loop of NaV1.8 to toxin activity. Results from this work may form the basis for future studies that lead to the rational design of spider venom-derived peptides with improved potency and selectivity at NaV1.8.

18.
Biomedicines ; 8(2)2020 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-32092883

RESUMEN

Spider venom is a novel source of disulfide-rich peptides with potent and selective activity at voltage-gated sodium channels (NaV). Here, we describe the discovery of µ-theraphotoxin-Pme1a and µ/δ-theraphotoxin-Pme2a, two novel peptides from the venom of the Gooty Ornamental tarantula Poecilotheria metallica that modulate NaV channels. Pme1a is a 35 residue peptide that inhibits NaV1.7 peak current (IC50 334 ± 114 nM) and shifts the voltage dependence of activation to more depolarised membrane potentials (V1/2 activation: Δ = +11.6 mV). Pme2a is a 33 residue peptide that delays fast inactivation and inhibits NaV1.7 peak current (EC50 > 10 µM). Synthesis of a [+22K]Pme2a analogue increased potency at NaV1.7 (IC50 5.6 ± 1.1 µM) and removed the effect of the native peptide on fast inactivation, indicating that a lysine at position 22 (Pme2a numbering) is important for inhibitory activity. Results from this study may be used to guide the rational design of spider venom-derived peptides with improved potency and selectivity at NaV channels in the future.

19.
Biochem Pharmacol ; 181: 114080, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32511987

RESUMEN

Management of chronic pain presents a major challenge, since many currently available treatments lack efficacy and have problems such as addiction and tolerance. Loss of function mutations in the SCN9A gene lead to a congenital inability to feel pain, with no other sensory deficits aside from anosmia. SCN9A encodes the voltage-gated sodium (NaV) channel 1.7 (NaV1.7), which has been identified as a primary pain target. However, in developing NaV1.7-targeted analgesics, extreme care must to be taken to avoid off-target activity on other NaV subtypes that are critical for survival. Since spider venoms are an excellent source of NaV channel modulators, we screened a panel of spider venoms to identify selective NaV1.7 inhibitors. This led to identification of two novel NaV modulating venom peptides (ß/µ-theraphotoxin-Pe1a and ß/µ-theraphotoxin-Pe1b (Pe1b) from the arboreal tarantula Phormingochilus everetti. A third peptide isolated from the tarantula Bumba pulcherrimaklaasi was identical to the well-known ProTx-I (ß/ω-theraphotoxin-Tp1a) from the tarantula Thrixopelma pruriens. A tethered toxin (t-toxin)-based alanine scanning strategy was used to determine the NaV1.7 pharmacophore of ProTx-I. We designed several ProTx-I and Pe1b analogues, and tested them for activity and NaV channel subtype selectivity. Several analogues had improved potency against NaV1.7, and altered specificity against other NaV channels. These analogues provide a foundation for development of Pe1b as a lead molecule for therapeutic inhibition of NaV1.7.


Asunto(s)
Analgésicos/farmacología , Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Péptidos/farmacología , Bloqueadores de los Canales de Sodio/farmacología , Secuencia de Aminoácidos , Aminoácidos/química , Aminoácidos/genética , Analgésicos/química , Analgésicos/aislamiento & purificación , Animales , Análisis Mutacional de ADN/métodos , Femenino , Humanos , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/genética , Activación del Canal Iónico/fisiología , Mutación , Canal de Sodio Activado por Voltaje NAV1.7/genética , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Oocitos/fisiología , Péptidos/química , Péptidos/genética , Conformación Proteica , Homología de Secuencia de Aminoácido , Bloqueadores de los Canales de Sodio/química , Bloqueadores de los Canales de Sodio/aislamiento & purificación , Venenos de Araña/química , Venenos de Araña/metabolismo , Xenopus laevis
20.
Biomedicines ; 8(7)2020 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-32629771

RESUMEN

Ant venoms have recently attracted increased attention due to their chemical complexity, novel molecular frameworks, and diverse biological activities. The heterodimeric peptide ∆-myrtoxin-Mp1a (Mp1a) from the venom of the Australian jack jumper ant, Myrmecia pilosula, exhibits antimicrobial, membrane-disrupting, and pain-inducing activities. In the present study, we examined the activity of Mp1a and a panel of synthetic analogues against the gastrointestinal parasitic nematode Haemonchus contortus, the fruit fly Drosophila melanogaster, and for their ability to stimulate pain-sensing neurons. Mp1a was found to be both insecticidal and anthelmintic, and it robustly activated mammalian sensory neurons at concentrations similar to those reported to elicit antimicrobial and cytotoxic activity. The native antiparallel Mp1a heterodimer was more potent than heterodimers with alternative disulfide connectivity, as well as monomeric analogues. We conclude that the membrane-disrupting effects of Mp1a confer broad-spectrum biological activities that facilitate both predation and defense for the ant. Our structure-activity data also provide a foundation for the rational engineering of analogues with selectivity for particular cell types.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA