Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Cancer Sci ; 104(9): 1226-30, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23718272

RESUMO

The marine snail peptide ziconotide (ω-conotoxin MVIIA) is used as an analgesic in cancer patients refractory to opioids, but may induce severe adverse effects. Animal venoms represent a rich source of novel drugs, so we investigated the analgesic effects and the side-effects of spider peptide Phα1ß in a model of cancer pain in mice with or without tolerance to morphine analgesia. Cancer pain was induced by the inoculation of melanoma B16-F10 cells into the hind paw of C57BL/6 mice. After 14 days, painful hypersensitivity was detected and Phα1ß or ω-conotoxin MVIIA (10-100 pmol/site) was intrathecally injected to evaluate the development of antinociception and side-effects in control and morphine-tolerant mice. The treatment with Phα1ß or ω-conotoxin MVIIA fully reversed cancer-related painful hypersensitivity, with long-lasting results, at effective doses 50% of 48 (32-72) or 33 (21-53) pmol/site, respectively. Phα1ß produced only mild adverse effects, whereas ω-conotoxin MVIIA induced dose-related side-effects in mice at analgesic doses (estimated toxic dose 50% of 30 pmol/site). In addition, we observed that Phα1ß was capable of controlling cancer-related pain even in mice tolerant to morphine antinociception (100% of inhibition) and was able to partially restore morphine analgesia in such animals (56 ± 5% of inhibition). In this study, Phα1ß was as efficacious as ω-conotoxin MVIIA in inducing analgesia in a model of cancer pain without producing severe adverse effects or losing efficacy in opioid-tolerant mice, indicating that Phα1ß has a good profile for the treatment of cancer pain in patients.


Assuntos
Analgésicos/farmacologia , Melanoma Experimental/tratamento farmacológico , Dor/tratamento farmacológico , Peptídeos/farmacologia , Venenos de Aranha/farmacologia , Aranhas/metabolismo , Analgésicos/efeitos adversos , Animais , Linhagem Celular Tumoral , Tolerância a Medicamentos , Masculino , Melanoma Experimental/complicações , Camundongos , Camundongos Endogâmicos C57BL , Morfina/efeitos adversos , Morfina/farmacologia , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Peptídeos/efeitos adversos , ômega-Conotoxinas/efeitos adversos , ômega-Conotoxinas/farmacologia
2.
Toxins (Basel) ; 15(9)2023 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-37755986

RESUMO

Diabetic neuropathic pain is one of the complications that affect a wide variety of the diabetic population and is often difficult to treat. Only a small number of patients experience pain relief, which usually comes with onerous side effects and low levels of satisfaction. The search for new analgesic drugs is necessary, given the limitations that current drugs present. Combining drugs to treat neuropathic pain has been attracting interest to improve their efficacy compared to single-drug monotherapies while also reducing dose sizes to minimize side effects. The aim of our study was to verify the antinociceptive effect of a synthetic peptide, PnPP-15, alone and combined with pregabalin, in male Swiss diabetic mice using the von Frey method. PnPP-15 is a synthetic peptide derived from PnPP19, a peptide representing a discontinuous epitope of the primary structure of the toxin PnTx2-6 from the venom of the spider Phoneutria nigriventer. The antinociceptive activity of both compounds was dose-dependent and showed synergism, which was verified by isobolographic analysis. Treatment with PnPP-15 did not cause spontaneous or forced motor changes and did not cause any damage or signs of toxicity in the analyzed organs (pancreas, lung, heart, kidney, brain, or liver). In conclusion, PnPP-15 is a great candidate for an analgesic drug against neuropathic pain caused by diabetes and exerts a synergistic effect when combined with pregabalin, allowing for even more efficient treatment.


Assuntos
Diabetes Mellitus Experimental , Neuralgia , Venenos de Aranha , Humanos , Ratos , Camundongos , Masculino , Animais , Pregabalina/farmacologia , Pregabalina/uso terapêutico , Ratos Wistar , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/tratamento farmacológico , Venenos de Aranha/uso terapêutico , Venenos de Aranha/toxicidade , Venenos de Aranha/química , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Analgésicos/farmacologia , Analgésicos/uso terapêutico , Neuralgia/tratamento farmacológico
3.
Curr Protein Pept Sci ; 24(5): 365-379, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37018532

RESUMO

Ion channels play critical roles in generating and propagating action potentials and in neurotransmitter release at a subset of excitatory and inhibitory synapses. Dysfunction of these channels has been linked to various health conditions, such as neurodegenerative diseases and chronic pain. Neurodegeneration is one of the underlying causes of a range of neurological pathologies, such as Alzheimer's disease (AD), Parkinson's disease (PD), cerebral ischemia, brain injury, and retinal ischemia. Pain is a symptom that can serve as an index of the severity and activity of a disease condition, a prognostic indicator, and a criterion of treatment efficacy. Neurological disorders and pain are conditions that undeniably impact a patient's survival, health, and quality of life, with possible financial consequences. Venoms are the best-known natural source of ion channel modulators. Venom peptides are increasingly recognized as potential therapeutic tools due to their high selectivity and potency gained through millions of years of evolutionary selection pressure. Spiders have been evolving complex and diverse repertoires of peptides in their venoms with vast pharmacological activities for more than 300 million years. These include peptides that potently and selectively modulate a range of targets, such as enzymes, receptors, and ion channels. Thus, components of spider venoms hold considerable capacity as drug candidates for alleviating or reducing neurodegeneration and pain. This review aims to summarize what is known about spider toxins acting upon ion channels, providing neuroprotective and analgesic effects.


Assuntos
Analgesia , Venenos de Aranha , Aranhas , Animais , Venenos de Aranha/farmacologia , Neuroproteção , Qualidade de Vida , Canais Iônicos , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Dor/tratamento farmacológico
4.
Mol Neurobiol ; 60(5): 2954-2968, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36754911

RESUMO

Some people living with HIV present painful sensory neuropathy (HIV-SN) that is pharmacoresistant, sex-associated, and a major source of morbidity. Since the specific mechanisms underlying HIV-SN are not well understood, the aim of our study was to characterize a novel model of painful HIV-SN by combining the HIV-1 gp120 protein and the antiretroviral stavudine (d4T) in mice and to investigate the pronociceptive role of the family 2 voltage-gated calcium channel (VGCC) α1 subunit (Cav2.X channels) in such a model. HIV-SN was induced in male and female C57BL/6 mice by administration of gp120 and/or d4T and detected by a battery of behavior tests and by immunohistochemistry. The role of Cav2.X channels was assessed by the treatment with selective blockers and agonists as well as by mRNA detection. Repeated administration with gp120 and/or d4T produced long-lasting touch-evoked painful-like behaviors (starting at 6 days, reaching a maximum on day 13, and lasting up to 28 days after treatment started), with a greater intensity in female mice treated with the combination of gp120 + d4T. Moreover, gp120 + d4T treatment reduced the intraepidermal nerve fibers and well-being of female mice, without altering other behaviors. Mechanistically, gp120 + d4T treatment induced Cav2.1, 2.2, and 2.3 transcriptional increases in the dorsal root ganglion and the Cav2.X agonist-induced nociception. Accordingly, intrathecal selective Cav2.2 blockade presented longer and better efficacy in reversing the hyperalgesia induced by gp120 + d4T treatment compared with Cav2.1 or Cav2.3, but also presented the worst safety (inducing side effects at effective doses). We conclude that the family 2 calcium channels (Cav2.X) exert a critical pronociceptive role in a novel mouse model of HIV-SN.


Assuntos
Dor Crônica , Infecções por HIV , Doenças do Sistema Nervoso Periférico , Masculino , Camundongos , Feminino , Animais , Estavudina/efeitos adversos , Camundongos Endogâmicos C57BL , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Canais de Cálcio Tipo N/metabolismo , Infecções por HIV/tratamento farmacológico , Dor Crônica/induzido quimicamente
5.
Artigo em Inglês | MEDLINE | ID: mdl-37205869

RESUMO

Introduction: Diabetic neuropathies are the most prevalent chronic complications of diabetes, characterized by pain and substantial morbidity. Although many drugs have been approved for the treatment of this type of pain, including gabapentin, tramadol (TMD), and classical opioids, it is common to report short-term results or potentially severe side effects. TMD, recommended as a second-line treatment can lead to unwanted side effects. Cannabidiol (CBD) has been gaining attention recently due to its therapeutic properties, including pain management. This study aimed to characterize the pharmacological interaction between CBD and TMD over the mechanical allodynia associated with experimental diabetes using isobolographic analysis. Materials and Methods: After diabetes induction by streptozotocin (STZ), diabetic rats were systemically treated with CBD or TMD alone or in combination (doses calculated based on linear regression of effective dose 40% [ED40]) and had the mechanical threshold evaluated using the electronic Von Frey apparatus. Both experimental and theoretical additive ED40 values (Zmix and Zadd, respectively) were determined for the combination of CBD plus TMD in this model. Results: Acute treatment with CBD (3 or 10 mg/kg) or TMD (2.5, 5, 10, or 20 mg/kg) alone or in combination (0.38+1.65 or 1.14+4.95 mg/kg) significantly improved the mechanical allodynia in STZ-diabetic rats. Isobolographic analysis revealed that experimental ED40 of the combination (Zmix) was 1.9 mg/kg (95% confidence interval [CI]=1.2-2.9) and did not differ from the theoretical additive ED40 2.0 mg/kg (95% CI=1.5-2.8; Zadd), suggesting an additive antinociceptive effect in this model. Conclusions: Using an isobolographic analysis, these results provide evidence of additive pharmacological interaction between CBD and TMD over the neuropathic pain associated with experimental diabetes induced by STZ.

6.
Front Mol Biosci ; 9: 876833, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35601827

RESUMO

Breast cancer is the most common cancer that affects women globally and is among the leading cause of women's death. Triple-negative breast cancer is more difficult to treat because hormone therapy is not available for this subset of cancer. The well-established therapy against triple-negative breast cancer is mainly based on surgery, chemotherapy, and immunotherapy. Among the drugs used in the therapy are cisplatin and carboplatin. However, they cause severe toxicity to the kidneys and brain and cause nausea. Therefore, it is urgent to propose new chemotherapy techniques that provide new treatment options to patients affected by this disease. Nowadays, peptide drugs are emerging as a class of promising new anticancer agents due to their lytic nature and, apparently, a minor drug resistance compared to other conventional drugs (reviewed in Jafari et al., 2022). We have recently reported the cytotoxic effect of the antimicrobial peptide LyeTx I-b against glioblastoma cells (Abdel-Salam et al., 2019). In this research, we demonstrated the cytotoxic effect of the peptide LyeTx I-b, alone and combined with cisplatin, against triple-negative cell lines (MDA-MD-231). LyeTx-I-b showed a selectivity index 70-fold higher than cisplatin. The peptide:cisplatin combination (P:C) 1:1 presented a synergistic effect on the cell death and a selective index value 16 times greater than the cisplatin alone treatment. Therefore, an equi-effective reduction of cisplatin can be reached in the presence of LyeTx I-b. Cells treated with P:C combinations were arrested in the G2/M cell cycle phase and showed positive staining for acridine orange, which was inhibited by bafilomycin A1, indicating autophagic cell death (ACD) as a probable cell death mechanism. Furthermore, Western blot experiments indicated a decrease in P21 expression and AKT phosphorylation. The decrease in AKT phosphorylation is indicative of ACD. However, other studies are still necessary to better elucidate the pathways involved in the cell death mechanism induced by the peptide and the drug combinations. These findings confirmed that the peptide LyeTx I-b seems to be a good candidate for combined chemotherapy to treat breast cancer. In addition, in vivo studies are essential to validate the use of LyeTx I-b as a therapeutic drug candidate, alone and/or combined with cisplatin.

7.
Artigo em Inglês | MEDLINE | ID: mdl-34512739

RESUMO

BACKGROUND: Phoneutria nigriventer venom contains Phα1ß. This toxin and its recombinant form have a remarkable analgesic potential that is associated with blockage of voltage-gated calcium channels and TRPA1 receptors. Although morphine is a mainstay drug to treat moderate and severe pain related to cancer, it has serious and dose-limiting side effects. Combining recombinant Phα1ß and morphine to treat pain is an interesting approach that has been gaining attention. Therefore, a quantitative and reliable method to establish the strength of the antinociceptive interaction between these two substances is necessary. The present study was designed to investigate the nature of the functional antinociceptive (analgesic) interaction between Phα1ß recombinant toxin and morphine in a model of cancer pain. METHODS: Melanoma was produced by intraplantar inoculation of B16-F10 cells into the right paw of C57BL/6J mice. Von Frey filaments measured the paw-withdrawal threshold after intrathecal administration of morphine, recombinant Phα1ß, and their combination. Thermal hyperalgesia was assessed using Hargreaves apparatus. The degree of interaction was evaluated using isobolographic analysis. Spontaneous and forced motor performance was assessed with the open-field and rotarod tests, respectively. RESULTS: Co-administration of recombinant Phα1ß and morphine synergistically reverses the melanoma-induced mechanical hyperalgesia. The potency of the mixture, measured as the effective dose to reach 50% of maximum possible effect (MPE) in ameliorating mechanical hyperalgesia, was about twice fold higher than expected if the interaction between morphine and recombinant Phα1ß was merely additive. Treatment with the combination at doses necessary to reach 50% of MPE caused no spontaneous nor forced motor alterations. CONCLUSION: The combinatorial use of recombinant Phα1ß and morphine allows significant and effective dose reduction of both agents, which has translational potential for opioid-sparing approaches in pain management related to cancer.

8.
Front Mol Biosci ; 8: 770471, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35187065

RESUMO

Preclinical evidence suggests the potential of Phα1ß, a toxin obtained from the venom of spider Phoneutria nigriventer, as a new analgesic drug. Molecular brain imaging techniques have afforded exciting opportunities to examine brain processes in clinical pain conditions. This paper aims to study the brain regions involved in the analgesic effects of Phα1ß compared with Morphine, in a model of acute pain induced by formalin in Sprague Dawley rats. We used 18F-fluorodeoxyglucose as a metabolic radiotracer to perform brain imaging of rats pretreated with Phα1ß or Morphine in a model of acute inflammatory pain caused by intraplantar injection of formalin. The rats' hind paw's formalin stimulation resulted in a brain metabolic increase at the bilateral motor cortex, visual cortex, somatosensory cortex, thalamus, and cingulate cortex.In rats treated with Phα1ß, selective inhibition of unilateral motor cortex and cingulate cortex was observed. Morphine treatment leads to small and selective inhibition at the bilateral amygdala striatum and accumbens. Our results indicate that the analgesic effect of Phα1ß and Morphine possesses a differential profile of central processing in the pain state.

9.
Biochem Pharmacol ; 182: 114210, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32882205

RESUMO

TRPV1 is a cation channel expressed in peripheral nociceptive pathways and its activation can trigger nociception signals to the brain. Ketamine is an intravenous anesthetic routinely used for anesthesia induction and with potent analgesic activity. Despite its proven depressant action on peripheral sensory pathways, the relationship between ketamine and TRPV1 receptors is still unclear. In this study, we evaluated the effect of ketamine injected peripherally in a rat model of spontaneous pain induced by capsaicin. We also investigated the effect of ketamine on Ca2+ transients in cultured dorsal root ganglia (DRG) neurons and HEK293 cells expressing the TRPV1 receptor (HEK-TRPV1 cells). Intraplantar administration of ketamine caused an unexpected increase in nocifensive behavior induced by capsaicin. Incubation of HEK-TRPV1 cells with 10 µM ketamine increased TRPV1 and PKCє phosphorylation. Ketamine potentiated capsaicin-induced Ca2+ transients in HEK-TRPV1 cells and DRG neurons. Ketamine also prevented TRPV1 receptor desensitization induced by successive applications of capsaicin. єV1-2, a PKCє inhibitor, reduced potentiation of capsaicin-induced Ca2+ transients by ketamine. Taken together, our data indicate that ketamine potentiates TRPV1 receptor sensitivity to capsaicin through a mechanism dependent on PKCє activity.


Assuntos
Ketamina/administração & dosagem , Nociceptividade/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Canais de Cátion TRPV/agonistas , Canais de Cátion TRPV/metabolismo , Animais , Capsaicina/administração & dosagem , Células Cultivadas , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Antagonistas de Aminoácidos Excitatórios/administração & dosagem , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Células HEK293 , Humanos , Masculino , Nociceptividade/fisiologia , Ratos , Ratos Wistar , Transdução de Sinais/fisiologia
10.
Pharmacol Rep ; 72(1): 47-54, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32016848

RESUMO

BACKGROUND: Diabetic neuropathy is a common cause of painful diabetic neuropathy (PDN). C-X-C chemokine receptor type 4 (CXCR4) expression is increased in peripheral nerve samples from diabetes patients, suggesting a role for CXCR4 in PDN. Therefore, we evaluated the effects of Phα1ß, ω-conotoxin MVIIA, and AMD3100 in a model of streptozotocin (STZ)-induced PDN in rodents and naïve model of rats with the activation of the CXCR4/stromal cell-derived factor 1 (SDF-1) signal. METHODS: Diabetic neuropathy was induced by intraperitoneal (ip) injection of STZ in Wistar rats. Naïve rats were intrathecally injected with SDF-1 to test the CXCR4/SDF-1 signal. The effects of Phα1ß intrathecal (it), ω-conotoxin MVIIA intrathecal (it), and AMD3100 intraperitoneal (ip) on rat hypersensitivity, IL-6, and the intracellular calcium [Ca2+]i content of diabetic synaptosomes were studied. RESULTS: The drugs reduced the hypersensitivity in diabetic rats. SDF-1 (1.0 µg/it) administration in naïve rats induced hypersensitivity. Phα1ß (100 pmol/it) or AMD3100 (2.5 µg/ip) reduced this hypersensitivity after 2 h treatments, while ω-conotoxin MVIIA did not have an effect. IL-6 and [Ca2+]i content increased in the spinal cord synaptosomes in diabetic rats. The drug treatments reduced IL-6 and the calcium influx in diabetic synaptosomes. CONCLUSIONS: Phα1ß, ω-conotoxin MVIIA, and AMD3100, after 2 h of treatment of STZ-induced PDN, reduced hypersensitivity in diabetic rats. In naïve rats with CXCR4/SDF-1 activation, the induced hypersensitivity decreased after 2 h treatments with Phα1ß or AMD-3100, while ω-conotoxin MVIIA did not affect. The inhibitory effects of Phα1ß on PDN may involve voltage-dependent calcium channels.


Assuntos
Analgésicos/farmacologia , Diabetes Mellitus Experimental/tratamento farmacológico , Neuropatias Diabéticas/tratamento farmacológico , Venenos de Aranha/farmacologia , Animais , Benzilaminas , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Quimiocina CXCL12/metabolismo , Ciclamos , Diabetes Mellitus Experimental/complicações , Compostos Heterocíclicos/farmacologia , Ratos , Ratos Wistar , Receptores CXCR4/metabolismo , ômega-Conotoxinas/farmacologia
11.
Neuropharmacology ; 162: 107826, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31647972

RESUMO

Capsaicin, an agonist of TRPV1, evokes intracellular [Ca2+] transients and glutamate release from perfused trigeminal ganglion. The spider toxin PnTx3-5, native or recombinant is more potent than the selective TRPV1 blocker SB-366791 with IC50 of 47 ±â€¯0.18 nM, 45 ±â€¯1.18 nM and 390 ±â€¯5.1 nM in the same experimental conditions. PnTx3-5 is thus more potent than the selective TRPV1 blocker SB-366791. PnTx3-5 (40 nM) and SB-366791 (3 µM) also inhibited the capsaicin-induced increase in intracellular Ca2+ in HEK293 cells transfected with TRPV1 by 75 ±â€¯16% and 84 ±â€¯3.2%, respectively. In HEK293 cells transfected with TRPA1, cinnamaldehyde (30 µM) generated an increase in intracellular Ca2+ that was blocked by the TRPA1 antagonist HC-030031 (10 µM, 89% inhibition), but not by PnTx3-5 (40 nM), indicating selectivity of the toxin for TRPV1. In whole-cell patch-clamp experiments on HEK293 cells transfected with TRPV1, capsaicin (10 µM) generated inward currents that were blocked by SB-366791 and by both native and recombinant PnTx3-5 by 47 ±â€¯1.4%; 54 ±â€¯7.8% and 56 ±â€¯9.0%, respectively. Intradermal injection of capsaicin into the rat left vibrissa induced nociceptive behavior that was blocked by pre-injection with either SB-366791 (3 nmol/site i.d., 83.3 ±â€¯7.2% inhibition) or PnTx3-5 (100 fmol/site, 89 ±â€¯8.4% inhibition). We conclude that both native and recombinant PnTx3-5 are potent TRPV1 receptor antagonists with antinociceptive action on pain behavior evoked by capsaicin.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Capsaicina/farmacologia , Dor Facial/metabolismo , Neuropeptídeos/farmacologia , Nociceptividade/efeitos dos fármacos , Fármacos do Sistema Sensorial/farmacologia , Canais de Cátion TRPV/antagonistas & inibidores , Gânglio Trigeminal/efeitos dos fármacos , Acroleína/análogos & derivados , Acroleína/farmacologia , Anilidas/farmacologia , Animais , Cálcio/metabolismo , Cinamatos/farmacologia , Modelos Animais de Doenças , Ácido Glutâmico/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Células HEK293 , Humanos , Concentração Inibidora 50 , Masculino , Técnicas de Patch-Clamp , Ratos , Canal de Cátion TRPA1/efeitos dos fármacos , Canal de Cátion TRPA1/genética , Canais de Cátion TRPV/genética , Transfecção , Gânglio Trigeminal/metabolismo
12.
Hippocampus ; 19(11): 1123-9, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19370546

RESUMO

The aim of this study was to investigate the effect of spider toxins on brain injury induced by oxygen deprivation and low glucose (ODLG) insult on slices of rat hippocampus. After ODLG insult cell viabilility in hippocampal slices was assessed by confocal microscopy and epifluorescence using the live/dead kit containing calcein-AM and ethidium homodimer and CA1 population spike amplitude recording during stimulation of Schaffer collateral fibers. Spider toxins Tx3-3 or Tx3-4 and conus toxins, omega-conotoxin GVIA or omega-conotoxin MVIIC are calcium channel blockers and protected against neuronal damage in slices subjected to ODLG insult. Confocal imaging of CA1 region of rat hippocampal slices subject to ischemic insult treated with Tx3-3, Tx3-4, omega-conotoxin GVIA or omega-conotoxin MVIIC showed a decrease in cell death that amounted to 68 +/- 4.2%, 77 +/- 3.8%, 32 +/- 2.3%, and 46 +/- 2.9%, respectively. This neuroprotective effect of Tx3-4 was corroborated by eletrophysiological recordings of population spikes amplitudes in CA1. The neuroprotection promoted on hippocampal slices by Tx3-3 or Tx3-4 was also observed when the toxins were applied 10, 20, 30, 60, 90, or 120 min after induction of the ODLG injury. During the ischemic insult, glutamate release from slices was increased by 71% (from 7.0 +/- 0.3 nM/mg of protein control slices not subjected to ischemia to 12 +/- 0.4 nM/mg of protein in slices exposed to ischemia). Tx3-3, Tx3-4, omega-conotoxin GVIA or omega-conotoxin MVIIC inhibited the ischemia-induced increase on glutamate release by 54, 72, 60, and 70%, respectively. Thus Tx3-3 and Tx3-4 provided robust ischemic neuroprotection showing potential as a novel class of agent that exerts neuroprotection in an in vitro model of brain ischemia.


Assuntos
Ácido Glutâmico/metabolismo , Hipocampo/patologia , Isquemia/patologia , Neurônios/efeitos dos fármacos , Venenos de Aranha/farmacologia , Transmissão Sináptica/efeitos dos fármacos , Análise de Variância , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Morte Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Hipocampo/efeitos dos fármacos , Técnicas In Vitro , Isquemia/tratamento farmacológico , Neuropeptídeos/farmacologia , Técnicas de Patch-Clamp/métodos , Ratos , Ratos Wistar , Fatores de Tempo , ômega-Conotoxina GVIA/farmacologia , ômega-Conotoxinas/farmacologia
13.
Neurosci Lett ; 439(2): 170-2, 2008 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-18524484

RESUMO

The purpose of the present work was to investigate the pharmacological action of a calcium channel-blocking toxin from the venom of the spider Phonetic nigriventer, Tx3-4 on calcium channels coupled to exocytosis of synaptic vesicles. Tx3-4 blocked KCl-induced exocytosis of synaptic vesicles with an IC50 of 1.1 nM. To investigate whether the target of Tx3-4 overlaps with known calcium channels that mediate calcium entry and exocytosis, we used omega-toxins that interact selectively with neuronal calcium channels. The results indicate that the main population of voltage-sensitive calcium channels altered by Tx3-4 is P/Q calcium channels. In conclusion, Tx3-4 is a potent inhibitor of calcium channels involved in the KCl-induced exocytosis of synaptic vesicles in brain cortical synaptosomes.


Assuntos
Canais de Cálcio/efeitos dos fármacos , Exocitose/efeitos dos fármacos , Neurotoxinas/farmacologia , Venenos de Aranha/farmacologia , Animais , Encéfalo/ultraestrutura , Bloqueadores dos Canais de Cálcio/farmacologia , Relação Dose-Resposta a Droga , Masculino , Neuropeptídeos/farmacologia , Cloreto de Potássio/farmacologia , Ratos , Ratos Wistar , Sinaptossomos/efeitos dos fármacos
14.
Scand J Pain ; 17: 203-210, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-29107209

RESUMO

BACKGROUND AND AIMS: Cholinergic agents cause antinociception by mimicking the release of acetylcholine (ACh) from spinal cholinergic nerves. PhKv is a peptide isolated from the venom of the armed spider Phoneutria nigriventer. It has an antiarrythmogenic activity that involves the enhanced release of acetylcholine. The aim of this study was to investigate whether PhKv had an antinociceptive action in mice. METHODS: Male albino Swiss mice (25-35g) were used in this study. The PhKv toxin was purified from a PhTx3 fraction of the Phoneutria nigriventer spider's venom. Because of its peptide nature, PhKv is not orally available and it was delivered directly into the central nervous system by an intrathecal (i.t.) route. PhKV on the thermal and mechanical sensitivity was evaluated using plantar test apparatus and the up-and-down method. The analgesic effects of PhKv were studied in neuropathic pain (CCI) and in the peripheral capsicin test. In order to test whether PhKv interfered with the cholinergic system, the mice were pre-treated with atropine (5mg/kg, i.p.) or mecamylamine (0.001mg/kg, i.p.) and the PhKv toxin (30pmol/site i.t.) or neostigmine (100pmol/site) were applied 15min before the intraplantar capsaicin (1nmol/paw) administrations. To investigate PhKv action on the AChE activities, was performed in vitro and ex vivo assay for AChE. For the in vitro experiments, mice spinal cord supernatants of tissue homogenates (1mg/ml) were used as source of AChE activity. The AChE assay was monitored at 37°C for 10min in a FlexStation 3 Multi-Mode Microplate Reader (Molecular Devices) at 405nm. RESULTS: PhKv (30 and 100pmol/site, i.t.) had no effect on the thermal or mechanical sensitivity thresholds. However, in a chronic constriction injury model of pain, PhKv (10pmol/site, i.t.) caused a robust reduction in mechanical withdrawal with an antinociceptive effect that lasted 4h. A pretreatment in mice with PhKv (30pmol/site, i.t.) or neostigmine (100pmol/site, i.t.) 15min before an intraplantar injection of capsaicin (1nmol/paw) caused a maximal antinociceptive effect of 69.5±4.9% and 85±2.5%, respectively. A pretreatment in mice with atropine; 5mg/kg, i.p. or mecamylamine 0.001mg/kg, i.p. inhibited a neostigimine and PhKv-induced antinociception, suggesting a cholinergic mechanism. Spinal acetylcholinesterase was inhibited by PhKv with ED50 of 7.6 (4.6-12.6pmol/site, i.t.). PhKv also inhibited the in vitro AChE activity of spinal cord homogenates with an EC50 of 20.8 (11.6-37.3nM), shifting the Km value from 0.06mM to 18.5mM, characterizing a competitive inhibition of AChE activity by PhKv. CONCLUSIONS: Our findings provide, to our knowledge, the first evidence that PhKv caused inhibition of AChE, it increased the ACh content at the neuronal synapses, leading to an activation of the cholinergic system and an antinociceptive response. IMPLICATIONS: Studies regarding the nociceptive mechanisms and the identification of potential targets for the treatment of pain have become top priorities. PhKv, by its action of stimulating the cholinergic receptors muscarinic and nicotinic system, reduces pain it may be an alternative for controlling the pain processes.


Assuntos
Analgésicos , Venenos de Aranha/química , Aranhas/química , Acetilcolina/metabolismo , Acetilcolina/fisiologia , Acetilcolinesterase/metabolismo , Analgésicos/administração & dosagem , Animais , Colinérgicos , Colinesterases , Técnicas In Vitro , Injeções Espinhais , Masculino , Camundongos , Dor/tratamento farmacológico , Venenos de Aranha/administração & dosagem
15.
Toxicon ; 133: 145-152, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28526335

RESUMO

The native Phα1ß - a Voltage-Gated Calcium Channel (VGCC) blocker - and its Recombinant Version - were both tested in rodent pain models with an intraplantar injections of capsaicin or formalin, a chronic constriction injury, and melanoma cancer related pain. The formalin nociceptive behaviour in the neurogenic phase was not affected by the toxin pre-treatments, while in the inflammatory phase, Phα1ß and the Recombinant form caused a significant reduction. The nociception that was triggered by capsaicin, an agonist of the TRPV1 vanilloid receptor, was totally blocked by 100 pmol/site, i.t. of Phα1ß or the recombinant version. For the neuropathic pain that was induced by a chronic constriction injury of the sciatic nerve, Phα1ß and its Recombinant reduced the allodynia that was induced by the CCI procedure in the rats and the hypersensitivity lasted for 4 h. Fourteen days after the inoculation of the B16-F10 melanoma cells in the mice, a marked hyperalgesia was induced in the melanoma cancer pain model. Phα1ß and the Recombinant form reduced the hyperalgesia with a full reversion at 100 pmol/site i.t. The inhibitory effects of the nociception that was induced by native Phα1ß and the Recombinant in the studied pain models were not statistically different and they developed with no side effects.


Assuntos
Analgésicos não Narcóticos/farmacologia , Neuralgia/tratamento farmacológico , Venenos de Aranha/farmacologia , Analgésicos não Narcóticos/uso terapêutico , Animais , Bloqueadores dos Canais de Cálcio , Capsaicina , Formaldeído , Masculino , Melanoma Experimental/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Dor Nociceptiva/tratamento farmacológico , Ratos Wistar , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Nervo Isquiático , Venenos de Aranha/uso terapêutico
16.
Artigo em Inglês | LILACS-Express | LILACS, VETINDEX | ID: biblio-1484780

RESUMO

Abstract Background: Phoneutria nigriventer venom contains Ph1. This toxin and its recombinant form have a remarkable analgesic potential that is associated with blockage of voltage-gated calcium channels and TRPA1 receptors. Although morphine is a mainstay drug to treat moderate and severe pain related to cancer, it has serious and dose-limiting side effects. Combining recombinant Ph1 and morphine to treat pain is an interesting approach that has been gaining attention. Therefore, a quantitative and reliable method to establish the strength of the antinociceptive interaction between these two substances is necessary. The present study was designed to investigate the nature of the functional antinociceptive (analgesic) interaction between Ph1 recombinant toxin and morphine in a model of cancer pain. Methods: Melanoma was produced by intraplantar inoculation of B16-F10 cells into the right paw of C57BL/6J mice. Von Frey filaments measured the paw-withdrawal threshold after intrathecal administration of morphine, recombinant Ph1, and their combination. Thermal hyperalgesia was assessed using Hargreaves apparatus. The degree of interaction was evaluated using isobolographic analysis. Spontaneous and forced motor performance was assessed with the open-field and rotarod tests, respectively. Results: Co-administration of recombinant Ph1 and morphine synergistically reverses the melanoma-induced mechanical hyperalgesia. The potency of the mixture, measured as the effective dose to reach 50% of maximum possible effect (MPE) in ameliorating mechanical hyperalgesia, was about twice fold higher than expected if the interaction between morphine and recombinant Ph1 was merely additive. Treatment with the combination at doses necessary to reach 50% of MPE caused no spontaneous nor forced motor alterations. Conclusion: The combinatorial use of recombinant Ph1 and morphine allows significant and effective dose reduction of both agents, which has translational potential for opioid-sparing approaches in pain management related to cancer.

17.
Toxins (Basel) ; 8(3)2016 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-26978403

RESUMO

The in vivo neuroprotective effect of PhTx3-4, a spider toxin N-P/Q calcium channel blocker, was studied in a rat model of NMDA-induced injury of the retina. NMDA (N-Methyl-D-Aspartate)-induced retinal injury in rats reduced the b-wave amplitude by 62% ± 3.6%, indicating the severity of the insult. PhTx3-4 treatment increased the amplitude of the b-wave, which was almost equivalent to the control retinas that were not submitted to injury. The PhTx3-4 functional protection of the retinas recorded on the ERG also was observed in the neuroprotection of retinal cells. NMDA-induced injury reduced live cells in the retina layers and the highest reduction, 84%, was in the ganglion cell layer. Notably, PhTx3-4 treatment caused a remarkable reduction of dead cells in the retina layers, and the highest neuroprotective effect was in the ganglion cells layer. NMDA-induced cytotoxicity of the retina increased the release of glutamate, reactive oxygen species (ROS) production and oxidative stress. PhTx3-4 treatment reduced glutamate release, ROS production and oxidative stress measured by malondialdehyde. Thus, we presented for the first time evidence of in vivo neuroprotection from NMDA-induced retinal injury by PhTx3-4 (-ctenitoxin-Pn3a), a spider toxin that blocks N-P/Q calcium channels.


Assuntos
Bloqueadores dos Canais de Cálcio/uso terapêutico , Neuropeptídeos/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Doenças Retinianas/tratamento farmacológico , Venenos de Aranha/uso terapêutico , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Eletrorretinografia , Ácido Glutâmico/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , N-Metilaspartato , Neuropeptídeos/farmacologia , Fármacos Neuroprotetores/farmacologia , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Doenças Retinianas/induzido quimicamente , Doenças Retinianas/metabolismo , Doenças Retinianas/fisiopatologia , Venenos de Aranha/farmacologia , Corpo Vítreo/metabolismo
18.
J. venom. anim. toxins incl. trop. dis ; 27: e20210001, 2021. tab, graf, ilus
Artigo em Inglês | LILACS, VETINDEX | ID: biblio-1484769

RESUMO

Phα1ß is a neurotoxin purified from spider venom that acts as a high-voltage-activated (HVA) calcium channel blocker. This spider peptide has shown a high selectivity for N-type HVA calcium channels (NVACC) and an analgesic effect in several animal models of pain. Its activity was associated with a reduction in calcium transients, glutamate release, and reactive oxygen species production from the spinal cord tissue and dorsal ganglia root (DRG) in rats and mice. It has been reported that intrathecal (i.t.) administration of Phα1ß to treat chronic pain reverted opioid tolerance with a safer profile than ω-conotoxin MVIIA, a highly selective NVACC blocker. Following a recent development of recombinant Phα1ß (CTK 01512-2), a new molecular target, TRPA1, the structural arrangement of disulphide bridges, and an effect on glial plasticity have been identified. CTK 01512-2 reproduced the antinociceptive effects of the native toxin not only after the intrathecal but also after the intravenous administration. Herein, we review the Phα1ß antinociceptive activity in the most relevant pain models and its mechanisms of action, highlighting the impact of CTK 01512-2 synthesis and its potential for multimodal analgesia.


Assuntos
Analgésicos/efeitos adversos , Dor , Espécies Reativas de Oxigênio , Neurotoxinas/isolamento & purificação , Peptídeos/isolamento & purificação
19.
Artigo em Inglês | LILACS-Express | LILACS, VETINDEX | ID: biblio-1484774

RESUMO

Abstract Ph1 is a neurotoxin purified from spider venom that acts as a high-voltage-activated (HVA) calcium channel blocker. This spider peptide has shown a high selectivity for N-type HVA calcium channels (NVACC) and an analgesic effect in several animal models of pain. Its activity was associated with a reduction in calcium transients, glutamate release, and reactive oxygen species production from the spinal cord tissue and dorsal ganglia root (DRG) in rats and mice. It has been reported that intrathecal (i.t.) administration of Ph1 to treat chronic pain reverted opioid tolerance with a safer profile than -conotoxin MVIIA, a highly selective NVACC blocker. Following a recent development of recombinant Ph1 (CTK 01512-2), a new molecular target, TRPA1, the structural arrangement of disulphide bridges, and an effect on glial plasticity have been identified. CTK 01512-2 reproduced the antinociceptive effects of the native toxin not only after the intrathecal but also after the intravenous administration. Herein, we review the Ph1 antinociceptive activity in the most relevant pain models and its mechanisms of action, highlighting the impact of CTK 01512-2 synthesis and its potential for multimodal analgesia.

20.
J. venom. anim. toxins incl. trop. dis ; 27: e20210001, 2021. tab, graf, ilus
Artigo em Inglês | LILACS, VETINDEX | ID: biblio-1351017

RESUMO

Phα1ß is a neurotoxin purified from spider venom that acts as a high-voltage-activated (HVA) calcium channel blocker. This spider peptide has shown a high selectivity for N-type HVA calcium channels (NVACC) and an analgesic effect in several animal models of pain. Its activity was associated with a reduction in calcium transients, glutamate release, and reactive oxygen species production from the spinal cord tissue and dorsal ganglia root (DRG) in rats and mice. It has been reported that intrathecal (i.t.) administration of Phα1ß to treat chronic pain reverted opioid tolerance with a safer profile than ω-conotoxin MVIIA, a highly selective NVACC blocker. Following a recent development of recombinant Phα1ß (CTK 01512-2), a new molecular target, TRPA1, the structural arrangement of disulphide bridges, and an effect on glial plasticity have been identified. CTK 01512-2 reproduced the antinociceptive effects of the native toxin not only after the intrathecal but also after the intravenous administration. Herein, we review the Phα1ß antinociceptive activity in the most relevant pain models and its mechanisms of action, highlighting the impact of CTK 01512-2 synthesis and its potential for multimodal analgesia.


Assuntos
Dor , Peptídeos/isolamento & purificação , Espécies Reativas de Oxigênio , Analgésicos/efeitos adversos , Neurotoxinas/isolamento & purificação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA