Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 8(1): 13397, 2018 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-30194442

RESUMO

Cone snails are a diverse group of predatory marine invertebrates that deploy remarkably complex venoms to rapidly paralyse worm, mollusc or fish prey. ω-Conotoxins are neurotoxic peptides from cone snail venoms that inhibit Cav2.2 voltage-gated calcium channel, demonstrating potential for pain management via intrathecal (IT) administration. Here, we isolated and characterized two novel ω-conotoxins, MoVIA and MoVIB from Conus moncuri, the first to be identified in vermivorous (worm-hunting) cone snails. MoVIA and MoVIB potently inhibited human Cav2.2 in fluorimetric assays and rat Cav2.2 in patch clamp studies, and both potently displaced radiolabeled ω-conotoxin GVIA (125I-GVIA) from human SH-SY5Y cells and fish brain membranes (IC50 2-9 pM). Intriguingly, an arginine at position 13 in MoVIA and MoVIB replaced the functionally critical tyrosine found in piscivorous ω-conotoxins. To investigate its role, we synthesized MoVIB-[R13Y] and MVIIA-[Y13R]. Interestingly, MVIIA-[Y13R] completely lost Cav2.2 activity and MoVIB-[R13Y] had reduced activity, indicating that Arg at position 13 was preferred in these vermivorous ω-conotoxins whereas tyrosine 13 is preferred in piscivorous ω-conotoxins. MoVIB reversed pain behavior in a rat neuropathic pain model, confirming that vermivorous cone snails are a new source of analgesic ω-conotoxins. Given vermivorous cone snails are ancestral to piscivorous species, our findings support the repurposing of defensive venom peptides in the evolution of piscivorous Conidae.


Assuntos
Analgésicos/química , Bloqueadores dos Canais de Cálcio/química , Evolução Molecular , ômega-Conotoxinas/química , Analgésicos/farmacologia , Analgésicos/uso terapêutico , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Bloqueadores dos Canais de Cálcio/uso terapêutico , Canais de Cálcio Tipo N/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Gânglios Espinais/citologia , Humanos , Neuralgia/tratamento farmacológico , Neurônios Aferentes/efeitos dos fármacos , Ratos , Ratos Wistar , Caramujos , ômega-Conotoxinas/genética , ômega-Conotoxinas/farmacologia , ômega-Conotoxinas/uso terapêutico
2.
PLoS One ; 12(9): e0182848, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28880874

RESUMO

Spider venoms are rich sources of peptidic ion channel modulators with important therapeutical potential. We screened a panel of 60 spider venoms to find modulators of ion channels involved in pain transmission. We isolated, synthesized and pharmacologically characterized Cd1a, a novel peptide from the venom of the spider Ceratogyrus darlingi. Cd1a reversibly paralysed sheep blowflies (PD50 of 1318 pmol/g) and inhibited human Cav2.2 (IC50 2.6 µM) but not Cav1.3 or Cav3.1 (IC50 > 30 µM) in fluorimetric assays. In patch-clamp electrophysiological assays Cd1a inhibited rat Cav2.2 with similar potency (IC50 3 µM) without influencing the voltage dependence of Cav2.2 activation gating, suggesting that Cd1a doesn't act on Cav2.2 as a classical gating modifier toxin. The Cd1a binding site on Cav2.2 did not overlap with that of the pore blocker ω-conotoxin GVIA, but its activity at Cav2.2-mutant indicated that Cd1a shares some molecular determinants with GVIA and MVIIA, localized near the pore region. Cd1a also inhibited human Nav1.1-1.2 and Nav1.7-1.8 (IC50 0.1-6.9 µM) but not Nav1.3-1.6 (IC50 > 30 µM) in fluorimetric assays. In patch-clamp assays, Cd1a strongly inhibited human Nav1.7 (IC50 16 nM) and produced a 29 mV depolarising shift in Nav1.7 voltage dependence of activation. Cd1a (400 pmol) fully reversed Nav1.7-evoked pain behaviours in mice without producing side effects. In conclusion, Cd1a inhibited two anti-nociceptive targets, appearing to interfere with Cav2.2 inactivation gating, associated with the Cav2.2 α-subunit pore, while altering the activation gating of Nav1.7. Cd1a was inactive at some of the Nav and Cav channels expressed in skeletal and cardiac muscles and nodes of Ranvier, apparently contributing to the lack of side effects at efficacious doses, and suggesting potential as a lead for development of peripheral pain treatments.


Assuntos
Analgésicos/farmacologia , Venenos de Aranha/química , Aranhas/química , Analgésicos/química , Animais , Sítios de Ligação/efeitos dos fármacos , Canais de Cálcio Tipo N/metabolismo , Eletrofisiologia , Fluorometria , Humanos , Camundongos , Canal de Sódio Disparado por Voltagem NAV1.1/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Peptídeos/química , Peptídeos/farmacologia , Ratos
3.
Toxins (Basel) ; 8(3)2016 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-26999206

RESUMO

Loss-of-function mutations of Na(V)1.7 lead to congenital insensitivity to pain, a rare condition resulting in individuals who are otherwise normal except for the inability to sense pain, making pharmacological inhibition of Na(V)1.7 a promising therapeutic strategy for the treatment of pain. We characterized a novel mouse model of Na(V)1.7-mediated pain based on intraplantar injection of the scorpion toxin OD1, which is suitable for rapid in vivo profiling of Na(V)1.7 inhibitors. Intraplantar injection of OD1 caused spontaneous pain behaviors, which were reversed by co-injection with Na(V)1.7 inhibitors and significantly reduced in Na(V)1.7(-/-) mice. To validate the use of the model for profiling Na(V)1.7 inhibitors, we determined the Na(V) selectivity and tested the efficacy of the reported Na(V)1.7 inhibitors GpTx-1, PF-04856264 and CNV1014802 (raxatrigine). GpTx-1 selectively inhibited Na(V)1.7 and was effective when co-administered with OD1, but lacked efficacy when delivered systemically. PF-04856264 state-dependently and selectively inhibited Na(V)1.7 and significantly reduced OD1-induced spontaneous pain when delivered locally and systemically. CNV1014802 state-dependently, but non-selectively, inhibited Na(V) channels and was only effective in the OD1 model when delivered systemically. Our novel model of Na(V)1.7-mediated pain based on intraplantar injection of OD1 is thus suitable for the rapid in vivo characterization of the analgesic efficacy of Na(V)1.7 inhibitors.


Assuntos
Canal de Sódio Disparado por Voltagem NAV1.7/fisiologia , Dor/tratamento farmacológico , Peptídeos/uso terapêutico , Éteres Fenílicos/uso terapêutico , Prolina/análogos & derivados , Venenos de Escorpião/uso terapêutico , Bloqueadores dos Canais de Sódio/uso terapêutico , Venenos de Aranha/uso terapêutico , Analgésicos , Animais , Comportamento Animal/efeitos dos fármacos , Células CHO , Cricetulus , Modelos Animais de Doenças , Células HEK293 , Humanos , Masculino , Camundongos Endogâmicos C57BL , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Fibras Nervosas/efeitos dos fármacos , Fibras Nervosas/fisiologia , Dor/induzido quimicamente , Prolina/uso terapêutico , Veia Safena/inervação , Sulfonamidas/uso terapêutico
4.
PLoS One ; 8(3): e59293, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23536870

RESUMO

SH-SY5Y human neuroblastoma cells provide a useful in vitro model to study the mechanisms underlying neurotransmission and nociception. These cells are derived from human sympathetic neuronal tissue and thus, express a number of the Cav channel subtypes essential for regulation of important physiological functions, such as heart contraction and nociception, including the clinically validated pain target Cav2.2. We have detected mRNA transcripts for a range of endogenous expressed subtypes Cav1.3, Cav2.2 (including two Cav1.3, and three Cav2.2 splice variant isoforms) and Cav3.1 in SH-SY5Y cells; as well as Cav auxiliary subunits α2δ1-3, ß1, ß3, ß4, γ1, γ4-5, and γ7. Both high- and low-voltage activated Cav channels generated calcium signals in SH-SY5Y cells. Pharmacological characterisation using ω-conotoxins CVID and MVIIA revealed significantly (∼ 10-fold) higher affinity at human versus rat Cav2.2, while GVIA, which interacts with Cav2.2 through a distinct pharmacophore had similar affinity for both species. CVID, GVIA and MVIIA affinity was higher for SH-SY5Y membranes vs whole cells in the binding assays and functional assays, suggesting auxiliary subunits expressed endogenously in native systems can strongly influence Cav2.2 channels pharmacology. These results may have implications for strategies used to identify therapeutic leads at Cav2.2 channels.


Assuntos
Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Expressão Gênica , Neuroblastoma/genética , Neuroblastoma/metabolismo , Ligação Competitiva , Bloqueadores dos Canais de Cálcio/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Humanos , Ligação Proteica , Isoformas de Proteínas , ômega-Conotoxina GVIA/metabolismo
5.
Toxins (Basel) ; 5(2): 286-314, 2013 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-23381143

RESUMO

Ca(v)2.2 is a calcium channel subtype localized at nerve terminals, including nociceptive fibers, where it initiates neurotransmitter release. Ca(v)2.2 is an important contributor to synaptic transmission in ascending pain pathways, and is up-regulated in the spinal cord in chronic pain states along with the auxiliary α2δ1 subunit. It is therefore not surprising that toxins that inhibit Ca(v)2.2 are analgesic. Venomous animals, such as cone snails, spiders, snakes, assassin bugs, centipedes and scorpions are rich sources of remarkably potent and selective Ca(v)2.2 inhibitors. However, side effects in humans currently limit their clinical use. Here we review Ca(v)2.2 inhibitors from venoms and their potential as drug leads.


Assuntos
Bloqueadores dos Canais de Cálcio/toxicidade , Canais de Cálcio Tipo N/fisiologia , Peptídeos/toxicidade , Peçonhas/química , Animais , Ensaios de Triagem em Larga Escala , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...