Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mar Drugs ; 20(10)2022 Sep 29.
Article in English | MEDLINE | ID: mdl-36286438

ABSTRACT

Neuroinflammation is a condition associated with several types of dementia, such as Alzheimer's disease (AD), mainly caused by an inflammatory response to amyloid peptides that induce microglial activation, with subsequent cytokine release. Neuronal caspase-1 from inflammasome and cathepsin B are key enzymes mediating neuroinflammation in AD, therefore, revealing new molecules to modulate these enzymes may be an interesting approach to treat neurodegenerative diseases. In this study, we searched for new caspase-1 and cathepsin B inhibitors from five species of Brazilian marine invertebrates (four cnidarians and one echinoderm). The results show that the extract of the box jellyfish Chiropsalmus quadrumanus inhibits caspase-1. This extract was fractionated, and the products monitored for their inhibitory activity, until the obtention of a pure molecule, which was identified as trigonelline by mass spectrometry. Moreover, four extracts inhibit cathepsin B, and Exaiptasia diaphana was selected for subsequent fractionation and characterization, resulting in the identification of betaine as being responsible for the inhibitory action. Both molecules are already found in marine organisms, however, this is the first study showing a potent inhibitory effect on caspase-1 and cathepsin B activities. Therefore, these new prototypes can be considered for the enzyme inhibition and subsequent control of the neuroinflammation.


Subject(s)
Alzheimer Disease , Cathepsin B , Humans , Animals , Caspase 1/pharmacology , Inflammasomes , Microglia , Neuroinflammatory Diseases , Aquatic Organisms , Betaine , Cytokines , Peptides/pharmacology , Invertebrates , Amyloid beta-Peptides/pharmacology
2.
Int J Mol Sci ; 23(19)2022 Sep 30.
Article in English | MEDLINE | ID: mdl-36232883

ABSTRACT

Pain is a worldwide public health problem and its treatment is still a challenge since clinically available drugs do not completely reverse chronic painful states or induce undesirable effects. Crotalphine is a 14 amino acids synthetic peptide that induces a potent and long-lasting analgesic effect on acute and chronic pain models, peripherally mediated by the endogenous release of dynorphin A and the desensitization of the transient receptor potential ankyrin 1 (TRPA1) receptor. However, the effects of crotalphine on the central nervous system (CNS) and the signaling pathway have not been investigated. Thus, the central effect of crotalphine was evaluated on the partial sciatic nerve ligation (PSNL)-induced chronic neuropathic pain model. Crotalphine (100 µg/kg, p.o.)-induced analgesia on the 14th day after surgery lasting up to 24 h after administration. This effect was prevented by intrathecal administration of CB1 (AM251) or CB2 (AM630) cannabinoid receptor antagonists. Besides that, crotalphine-induced analgesia was reversed by CTOP, nor-BNI, and naltrindole, antagonists of mu, kappa, and delta-opioid receptors, respectively, and also by the specific antibodies for ß-endorphin, dynorphin-A, and met-enkephalin. Likewise, the analgesic effect of crotalphine was blocked by the intrathecal administration of minocycline, an inhibitor of microglial activation and proliferation. Additionally, crotalphine decreased the PSNL-induced IL-6 release in the spinal cord. Importantly, in vitro, crotalphine inhibited LPS-induced CD86 expression and upregulated CD206 expression in BV-2 cells, demonstrating a polarization of microglial cells towards the M2 phenotype. These results demonstrated that crotalphine, besides activating opioid and cannabinoid analgesic systems, impairs central neuroinflammation, confirming the neuromodulatory mechanism involved in the crotalphine analgesic effect.


Subject(s)
Analgesia , Cannabinoids , Neuralgia , Amino Acids/metabolism , Analgesics/metabolism , Analgesics/pharmacology , Analgesics/therapeutic use , Analgesics, Opioid/metabolism , Ankyrins/metabolism , Cannabinoid Receptor Antagonists/therapeutic use , Cannabinoids/therapeutic use , Dynorphins/metabolism , Enkephalin, Methionine/metabolism , Humans , Interleukin-6/metabolism , Lipopolysaccharides/metabolism , Microglia/metabolism , Minocycline/therapeutic use , Neuralgia/metabolism , Peptides , Phenotype , Receptors, Opioid/metabolism , Spinal Cord , beta-Endorphin/metabolism
3.
Blood Cells Mol Dis ; 92: 102604, 2021 12.
Article in English | MEDLINE | ID: mdl-34517295

ABSTRACT

Patients with COVID-19 can be asymptomatic or present mild to severe symptoms, leading to respiratory and cardiovascular complications and death. Type 2 diabetes mellitus (T2DM) and obesity are considered risk factors for COVID-19 poor prognosis. In parallel, COVID-19 severe patients exhibit dyslipidemia and alterations in neutrophil to lymphocyte ratio (NLR) associated with disease severity and mortality. To investigate whether such alterations are caused by the infection or results from preexisting comorbidities, this work analyzed dyslipidemia and the hemogram profile of COVID-19 patients according to the severity and compared with patients without T2DM or obesity comorbidities. Dyslipidemia, with a marked decrease in HDL levels, and increased NLR accompanied the disease severity, even in non-T2DM and non-obese patients, indicating that COVID-19 causes the observed alterations. Because decreased hemoglobin is involved in COVID-19 severity, and hemoglobin concentration is associated with metabolic diseases, the erythrogram of patients was also evaluated. We verified a drop in hemoglobin and erythrocyte number in severe patients, independently of T2DM and obesity, which may explain in part the need for artificial ventilation in severe cases. Thus, the control of such parameters (especially HDL levels, NLR, and hemoglobin concentration) could be a good strategy to prevent COVID-19 complications and death.


Subject(s)
Atherosclerosis/etiology , COVID-19/complications , Dyslipidemias/etiology , Leukocyte Count , SARS-CoV-2 , Adult , Aged , Anemia/epidemiology , Anemia/etiology , Atherosclerosis/epidemiology , COVID-19/blood , COVID-19/therapy , Comorbidity , Diabetes Mellitus, Type 2/epidemiology , Dyslipidemias/epidemiology , Erythrocyte Count , Hemoglobins/analysis , Humans , Hypoxia/etiology , Hypoxia/therapy , Lipoproteins, HDL/blood , Lymphocyte Count , Middle Aged , Neutrophils , Obesity/epidemiology , Respiration, Artificial , Retrospective Studies , Risk Factors , Severity of Illness Index
4.
J Neuroinflammation ; 17(1): 266, 2020 Sep 07.
Article in English | MEDLINE | ID: mdl-32894170

ABSTRACT

BACKGROUND: Experimental autoimmune encephalomyelitis (EAE) is the most commonly used and clinically relevant murine model for human multiple sclerosis (MS), a demyelinating autoimmune disease characterized by mononuclear cell infiltration into the central nervous system (CNS). The aim of the present study was to appraise the alterations, poorly documented in the literature, which may occur at the peripheral nervous system (PNS) level. METHODS: To this purpose, a multiple evaluation of peripheral nerve excitability was undertaken, by means of a minimally invasive electrophysiological method, in EAE mice immunized with the myelin oligodendrocyte glycoprotein (MOG) 35-55 peptide, an experimental model for MS that reproduces, in animals, the anatomical and behavioral alterations observed in humans with MS, including CNS inflammation, demyelination of neurons, and motor abnormalities. Additionally, the myelin sheath thickness of mouse sciatic nerves was evaluated using transmission electronic microscopy. RESULTS: As expected, the mean clinical score of mice, daily determined to describe the symptoms associated to the EAE progression, increased within about 18 days after immunization for EAE mice while it remained null for all control animals. The multiple evaluation of peripheral nerve excitability, performed in vivo 2 and 4 weeks after immunization, reveals that the main modifications of EAE mice, compared to control animals, are a decrease of the maximal compound action potential (CAP) amplitude and of the stimulation intensity necessary to generate a CAP with a 50% maximum amplitude. In addition, and in contrast to control mice, at least 2 CAPs were recorded following a single stimulation in EAE animals, reflecting various populations of sensory and motor nerve fibers having different CAP conduction speeds, as expected if a demyelinating process occurred in the PNS of these animals. In contrast, single CAPs were always recorded from the sensory and motor nerve fibers of control mice having more homogeneous CAP conduction speeds. Finally, the myelin sheath thickness of sciatic nerves of EAE mice was decreased 4 weeks after immunization when compared to control animals. CONCLUSIONS: In conclusion, the loss of immunological self-tolerance to MOG in EAE mice or in MS patients may not be only attributed to the restricted expression of this antigen in the immunologically privileged environment of the CNS but also of the PNS.


Subject(s)
Action Potentials/physiology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Neural Conduction/physiology , Peripheral Nerves/physiopathology , Animals , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Myelin Sheath/immunology , Myelin Sheath/pathology , Myelin-Oligodendrocyte Glycoprotein/immunology , Peptide Fragments/immunology , Peripheral Nerves/immunology , Peripheral Nerves/pathology
5.
Phys Life Rev ; 48: 176-197, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38320380

ABSTRACT

It is more than recognized and accepted that the environment affects the physiological responses of all living things, from bacteria to superior vertebrates, constituting an important factor in the evolution of all species. Environmental influences range from natural processes such as sunlight, seasons of the year, and rest to complex processes like stress and other mood disorders, infections, and air pollution, being all of them influenced by how each creature deals with them. In this chapter, it will be discussed how some of the environmental elements affect directly or indirectly neuropathic pain, a type of chronic pain caused by a lesion or disease of the somatosensory nervous system. For that, it was considered the edge of knowledge in translational research, thus including data from human and experimental animals as well as the applicability of such findings.


Subject(s)
Air Pollution , Chronic Pain , Neuralgia , Humans , Animals , Chronic Pain/complications , Neuralgia/etiology , Seasons
7.
Toxicon ; 238: 107568, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38110040

ABSTRACT

Most anti-inflammatory drugs currently adopted to treat chronic inflammatory joint diseases can alleviate symptoms but they do not lead to remission. Therefore, new and more efficient drugs are needed to block the course of joint inflammatory diseases. Animal venoms, rich in bioactive compounds, can contribute as valuable tools in this field of research. In this study, we first demonstrate the direct action of venoms on cells that constitute the articular joints. We established a platform consisting of cell-based assays to evaluate the release of cytokines (IL-6, IL-8, TNFα, IL-1ß, and IL-10) by human chondrocytes, synoviocytes and THP1 macrophages, as well as the release of neuropeptides (substance-P and ß-endorphin) by differentiated sensory neuron-like cells, 24 h after stimulation of cells with 21 animal venoms from snake and arthropod species, sourced from different taxonomic families and geographic origins. Results demonstrated that at non-cytotoxic concentrations, the venoms activate at varying degrees the secretion of inflammatory mediators involved in the pathology of articular diseases, such as IL-6, IL-8, and TNF-α by chondrocytes, synoviocytes, and macrophages and of substance P by neuron-like cells. Venoms of the Viperidae snake family were more inflammatory than those of the Elapidae family, while venoms of Arthropods were less inflammatory than snake venoms. Notably, some venoms also induced the release of the anti-inflammatory IL-10 by macrophages. However, the scorpion Buthus occitanus venom induced the release of IL-10 without increasing the release of inflammatory cytokines by macrophages. Since the cell types used in the experiments are crucial elements in joint inflammatory processes, the results of this work may guide future research on the activation of receptors and inflammatory signaling pathways by selected venoms in these particular cells, aiming at discovering new targets for therapeutic intervention.


Subject(s)
Animals, Poisonous , Arthropod Venoms , Arthropods , Joint Diseases , Scorpion Venoms , Scorpions , Viperidae , Animals , Humans , Interleukin-10 , Interleukin-6 , Interleukin-8 , Snake Venoms/chemistry , Cytokines , Tumor Necrosis Factor-alpha , Anti-Inflammatory Agents
8.
Int J Biol Macromol ; 242(Pt 2): 124892, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37196721

ABSTRACT

Cancer is the second leading cause of death worldwide, and despite the effort of standard treatments, the search for new tools against this disease is necessary. Importantly, it is known that the tumor microenvironment plays a crucial role in tumor initiation, progression, and response to therapies. Therefore, studies of potential drugs that act on these components are as critical as studies regarding antiproliferative substances. Through the years, studies of several natural products, including animal toxins, have been conducted to guide the development of medical compounds. In this review, we present the remarkable antitumor activities of crotoxin, a toxin from the rattlesnake Crotalus durissus terrificus, highlighting its effects on cancer cells and in the modulation of relevant elements in the tumor microenvironment as well as the clinical trials conducted with this compound. In summary, crotoxin acts through several mechanisms of action, such as activation of apoptosis, induction of cell cycle arrest, inhibition of metastasis, and decrease of tumor growth, in different tumor types. Crotoxin also modulates tumor-associated fibroblasts, endothelial cells, and immune cells, which contribute to its antitumoral effects. In addition, preliminary clinical studies confirm the promising results of crotoxin and support its potential future use as an anticancer drug.


Subject(s)
Antineoplastic Agents , Crotalid Venoms , Crotoxin , Neoplasms , Animals , Crotoxin/pharmacology , Crotalid Venoms/toxicity , Endothelial Cells/metabolism , Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Tumor Microenvironment
9.
Behav Pharmacol ; 23(1): 14-24, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22126967

ABSTRACT

Crotalphine, a 14 amino acid peptide first isolated from the venom of the South American rattlesnake Crotalus durissus terrificus, induces a peripheral long-lasting and opioid receptor-mediated antinociceptive effect in a rat model of neuropathic pain induced by chronic constriction of the sciatic nerve. In the present study, we further characterized the molecular mechanisms involved in this effect, determining the type of opioid receptor responsible for this effect and the involvement of the nitric oxide-cyclic GMP pathway and of K⁺ channels. Crotalphine (0.2 or 5 µg/kg, orally; 0.0006 µg/paw), administered on day 14 after nerve constriction, inhibited mechanical hyperalgesia and low-threshold mechanical allodynia. The effect of the peptide was antagonized by intraplantar administration of naltrindole, an antagonist of δ-opioid receptors, and partially reversed by norbinaltorphimine, an antagonist of κ-opioid receptors. The effect of crotalphine was also blocked by 7-nitroindazole, an inhibitor of the neuronal nitric oxide synthase; by 1H-(1,2,4) oxadiazolo[4,3-a]quinoxaline-1-one, an inhibitor of guanylate cyclase activation; and by glibenclamide, an ATP-sensitive K⁺ channel blocker. The results suggest that peripheral δ-opioid and κ-opioid receptors, the nitric oxide-cyclic GMP pathway, and ATP-sensitive K⁺ channels are involved in the antinociceptive effect of crotalphine. The present data point to the therapeutic potential of this peptide for the treatment of chronic neuropathic pain.


Subject(s)
Analgesics/pharmacology , Arginine/physiology , Cyclic GMP/physiology , KATP Channels/physiology , Neuralgia/drug therapy , Nitric Oxide/physiology , Peptides/pharmacology , Animals , Male , Rats , Rats, Wistar , Receptors, Opioid, delta/physiology , Receptors, Opioid, kappa/physiology , Signal Transduction/physiology
10.
Br J Pharmacol ; 179(8): 1640-1660, 2022 04.
Article in English | MEDLINE | ID: mdl-34076891

ABSTRACT

Depression and anxiety commonly occur in chronic pain states and the coexistence of these diseases worsens outcomes for both disorders and may reduce treatment adherence and response. Despite the advances in the knowledge of chronic pain mechanisms, pharmacological treatment is still unsatisfactory. Research based on exposure to environmental enrichment is currently under investigation and seems to offer a promising low-cost strategy with no side effects. In this review, we discuss the role of inflammation as a major biological substrate and aetiological factor of chronic pain and depression/anxiety and report a collection of preclinical evidence of the effects and mechanisms of environmental enrichment. As microglia participates in the development of both conditions, we also discuss microglia as a potential target underlying the beneficial actions of environmental enrichment in chronic pain and comorbid depression/anxiety. We also discuss how alternative interventions under clinical guidelines, such as environmental enrichment, may improve treatment compliance and patient outcomes. LINKED ARTICLES: This article is part of a themed issue on Building Bridges in Neuropharmacology. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v179.8/issuetoc.


Subject(s)
Chronic Pain , Anxiety Disorders/therapy , Chronic Pain/drug therapy , Depression/drug therapy , Humans , Neuroinflammatory Diseases , Neuropharmacology
11.
Cells ; 11(2)2022 01 12.
Article in English | MEDLINE | ID: mdl-35053363

ABSTRACT

Increased collagen-derived advanced glycation end-products (AGEs) are consistently related to painful diseases, including osteoarthritis, diabetic neuropathy, and neurodegenerative disorders. We have recently developed a model combining a two-dimensional glycated extracellular matrix (ECM-GC) and primary dorsal root ganglion (DRG) that mimicked a pro-nociceptive microenvironment. However, culturing primary cells is still a challenge for large-scale screening studies. Here, we characterized a new model using ECM-GC as a stimulus for human sensory-like neurons differentiated from SH-SY5Y cell lines to screen for analgesic compounds. First, we confirmed that the differentiation process induces the expression of neuron markers (MAP2, RBFOX3 (NeuN), and TUBB3 (ß-III tubulin), as well as sensory neuron markers critical for pain sensation (TRPV1, SCN9A (Nav1.7), SCN10A (Nav1.8), and SCN11A (Nav1.9). Next, we showed that ECM-GC increased c-Fos expression in human sensory-like neurons, which is suggestive of neuronal activation. In addition, ECM-GC upregulated the expression of critical genes involved in pain, including SCN9A and TACR1. Of interest, ECM-GC induced substance P release, a neuropeptide widely involved in neuroinflammation and pain. Finally, morphine, the prototype opiate, decreased ECM-GC-induced substance P release. Together, our results suggest that we established a functional model that can be useful as a platform for screening candidates for the management of painful conditions.


Subject(s)
Analgesics/analysis , Analgesics/pharmacology , Collagen/pharmacology , Drug Evaluation, Preclinical , Models, Biological , Sensory Receptor Cells/cytology , Animals , Antigens, Neoplasm/metabolism , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Extracellular Matrix/metabolism , Galectin 3/metabolism , Gene Expression Regulation/drug effects , Glycosylation/drug effects , Humans , Mitogen-Activated Protein Kinases/metabolism , NAV1.7 Voltage-Gated Sodium Channel/genetics , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Neurites/drug effects , Neurites/metabolism , Neurons/cytology , Neurons/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Rats , Receptors, Neurokinin-1/genetics , Receptors, Neurokinin-1/metabolism , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Substance P/metabolism , beta-Endorphin/metabolism
12.
Nitric Oxide ; 25(3): 243-54, 2011 Oct 30.
Article in English | MEDLINE | ID: mdl-21723953

ABSTRACT

Nitric oxide (NO) is involved in many physiological processes and several lines of evidence have indicated that NO plays a complex and diverse role in the modulation of pain. Nitric oxide is an important neurotransmitter involved in the nociceptive process and, in the dorsal horn of the spinal cord, it contributes to the development of central sensitization. On the other hand, experimental data have also demonstrated that NO inhibits nociception in the peripheral and also in the central nervous system. In addition, it has been shown that nitric oxide mediates the analgesic effect of opioids and other analgesic substances. The information included in the present review aims to present and analyze data about the dual effect of NO on pain transmission and control, the molecular mechanisms involved in these effects and also the potential use of nitric oxide in pain therapy.


Subject(s)
Analgesia/methods , Nitric Oxide/metabolism , Nociceptors/metabolism , Pain/metabolism , Animals , Humans , Nitric Oxide/therapeutic use , Pain/drug therapy , Pain/physiopathology , Pain Measurement
13.
J Nat Prod ; 74(3): 378-82, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21309590

ABSTRACT

A new acylamino acid, bunodosine 391 (BDS 391), was isolated from the venom of the sea anemone Bunodosoma cangicum. The structure was elucidated by spectroscopic analyses (2D NMR, ESIMS/MS) and verified by its synthesis. Intraplantar injection of BDS 391 into the hind paw of a rat induced a potent analgesic effect. This effect was not altered by naloxone (an opioid receptor antagonist), but was completely reversed by methysergide (a serotonin receptor antagonist), indicating that the effect is mediated by activation of serotonin receptors.


Subject(s)
Analgesics/isolation & purification , Analgesics/pharmacology , Sea Anemones/chemistry , Analgesics/chemistry , Animals , Cnidarian Venoms/chemical synthesis , Cnidarian Venoms/chemistry , Cnidarian Venoms/isolation & purification , Cnidarian Venoms/pharmacology , Edema/chemically induced , Edema/drug therapy , Hindlimb/drug effects , Male , Molecular Structure , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Nuclear Magnetic Resonance, Biomolecular , Rats , Rats, Wistar , Receptors, Serotonin/drug effects , Receptors, Serotonin/metabolism
14.
Toxins (Basel) ; 13(12)2021 12 20.
Article in English | MEDLINE | ID: mdl-34941749

ABSTRACT

Crotalphine (CRP) is a structural analogue to a peptide that was first identified in the crude venom from the South American rattlesnake Crotalus durissus terrificus. This peptide induces a potent and long-lasting antinociceptive effect that is mediated by the activation of peripheral opioid receptors. The opioid receptor activation regulates a variety of intracellular signaling, including the mitogen-activated protein kinase (MAPK) pathway. Using primary cultures of sensory neurons, it was demonstrated that crotalphine increases the level of activated ERK1/2 and JNK-MAPKs and this increase is dependent on the activation of protein kinase Cζ (PKCζ). However, whether PKCζ-MAPK signaling is critical for crotalphine-induced antinociception is unknown. Here, we biochemically demonstrated that the systemic crotalphine activates ERK1/2 and JNK and decreases the phosphorylation of p38 in the lumbar spinal cord. The in vivo pharmacological inhibition of spinal ERK1/2 and JNK, but not of p38, blocks the antinociceptive effect of crotalphine. Of interest, the administration of a PKCζ pseudosubstrate (PKCζ inhibitor) prevents crotalphine-induced ERK activation in the spinal cord, followed by the abolishment of crotalphine-induced analgesia. Together, our results demonstrate that the PKCζ-ERK signaling pathway is involved in crotalphine-induced analgesia. Our study opens a perspective for the PKCζ-MAPK axis as a target for pain control.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Pain/drug therapy , Peptides/pharmacology , Protein Kinase C/metabolism , Signal Transduction/drug effects , Animals , Behavior, Animal , Gene Expression Regulation, Enzymologic/drug effects , Mitogen-Activated Protein Kinases/genetics , Protein Kinase C/genetics , Rats , Rats, Wistar
15.
Biomed Res Int ; 2021: 8855248, 2021.
Article in English | MEDLINE | ID: mdl-33748281

ABSTRACT

Neurodegenerative diseases are one of the major causes of death worldwide, characterized by neurite atrophy, neuron apoptosis, and synapse loss. No effective treatment has been indicated for such diseases so far, and the search for new drugs is being increased in the last years. Animal venoms' secretion/venom can be an alternative for the discovery of new molecules, which could be the prototype for a new treatment. Here, we present the biochemical characterization and activity of the extract from the box jellyfish Chiropsalmus quadrumanus (Cq) on neurites. The Cq methanolic extract was obtained and incubated to human SH-SY5Y neurons, and neurite parameters were evaluated. The extract was tested in other cell types to check its cytotoxicity and was submitted to biochemical analysis by mass spectrometry in order to check its composition. We could verify that the Cq extract increased neurite outgrowth length and branching junctions, amplifying the contact between SH-SY5Y neurons, without affecting cell body and viability. The extract action was selective for neurons, as it did not cause any effects on other cell types, such as tumor line, nontumor line, and red blood cells. Moreover, mass spectrometry analysis revealed that there are no proteins but several low molecular mass compounds and peptides. Three peptides, characterized as cryptides, and 14 low molecular mass compounds were found to be related to cytoskeleton reorganization, cell membrane expansion, and antioxidant/neuroprotective activity, which act together to increase neuritogenesis. After this evaluation, we conclude that the Cq extract is a promising tool for neuronal connection recovery, an essential condition for the treatment of neurodegenerative diseases.


Subject(s)
Complex Mixtures/pharmacology , Cubozoa/chemistry , Neurites/metabolism , Neuroprotective Agents/pharmacology , Animals , Cell Line, Tumor , Complex Mixtures/chemistry , Humans , Neuroprotective Agents/chemistry
16.
Toxins (Basel) ; 13(11)2021 11 22.
Article in English | MEDLINE | ID: mdl-34822611

ABSTRACT

Multiple sclerosis (MS) is a demyelinating disease of inflammatory and autoimmune origin, which induces sensory and progressive motor impairments, including pain. Cells of the immune system actively participate in the pathogenesis and progression of MS by inducing neuroinflammation, tissue damage, and demyelination. Crotalphine (CRO), a structural analogue to a peptide firstly identified in Crotalus durissus terrificus snake venom, induces analgesia by endogenous opioid release and type 2 cannabinoid receptor (CB2) activation. Since CB2 activation downregulates neuroinflammation and ameliorates symptoms in mice models of MS, it was presently investigated whether CRO has a beneficial effect in the experimental autoimmune encephalomyelitis (EAE). CRO was administered on the 5th day after immunization, in a single dose, or five doses starting at the peak of disease. CRO partially reverted EAE-induced mechanical hyperalgesia and decreased the severity of the clinical signs. In addition, CRO decreases the inflammatory infiltrate and glial cells activation followed by TNF-α and IL-17 downregulation in the spinal cord. Peripherally, CRO recovers the EAE-induced impairment in myelin thickness in the sciatic nerve. Therefore, CRO interferes with central and peripheral neuroinflammation, opening perspectives to MS control.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Neuroinflammatory Diseases/drug therapy , Pain/drug therapy , Peptides/pharmacology , Analgesics/pharmacology , Animals , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Hyperalgesia/drug therapy , Mice , Mice, Inbred C57BL , Multiple Sclerosis/drug therapy , Multiple Sclerosis/physiopathology , Receptor, Cannabinoid, CB2/drug effects , Receptor, Cannabinoid, CB2/metabolism
17.
Front Immunol ; 12: 779473, 2021.
Article in English | MEDLINE | ID: mdl-35185861

ABSTRACT

Bothrops jararaca venom (BjV) can induce mast cell degranulation. In order to investigate the role of mast cells and the interference of the host genetic background in the inflammation induced by BjV, we have used mouse strains selected for maximal (AIRmax) or minimal (AIRmin) acute inflammatory response (AIR). Mice were pretreated with an inhibitor of mast cell degranulation, cromolyn (CROM), and injected in footpads or intraperitoneally (i.p.) with BjV. Pain was measured with von Frey hairs, cell migration in the peritoneum by flow cytometry, and reactive oxygen species (ROS) production by chemiluminescence assays. The nociceptive response to BjV was higher in AIRmax than AIRmin mice; however, this difference was abolished by pretreatment with CROM. BjV induced peritoneal neutrophil (CD11b+ GR-1+) infiltration and ROS secretion in AIRmax mice only, which were partially inhibited by CROM. Our findings evidence a role for mast cells in pain, neutrophil migration, and ROS production triggered by BjV in AIRmax mice that are more susceptible to the action of BjV.


Subject(s)
Bothrops , Crotalid Venoms , Animals , Cell Movement , Crotalid Venoms/adverse effects , Inflammation/chemically induced , Mast Cells , Mice , Pain , Reactive Oxygen Species
18.
Toxicon ; 199: 127-138, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34139257

ABSTRACT

Cnidarians are equipped with nematocysts, which are specialized organelles used to inoculate venom during prey capturing and defense. Their venoms are rich in toxins and a potential source of bioactive compounds, however, poorly explored so far. In this work, the activity of the methanolic extracts from the hydromedusa Olindias sambaquiensis and the cubozoan jellyfish Chiropsalmus quadrumanus were studied in sympathetic neurotransmission. For that, bisected rat vas deferens - a classic model of sympathetic neurotransmission - were incubated with the extracts for further myographic and histopathological analysis. The O. sambaquiensis extract, at 0.1 µg/mL, facilitated the neurogenic contractions of the noradrenergic-rich epididymal portion, while reducing the noradrenaline (NA) potency, which suggests an interaction with postsynaptic α1-adrenoceptors. On the other hand, a higher concentration (1 µg/mL) leads to time- and frequency-dependent blockade of nerve-evoked contractions without significantly changing the response to exogenous NA. In turn, the C. quadrumanus extract at 0.1 µg/mL induced blockade of nerve-evoked noradrenergic contractions while reducing the potency to exogenous NA. Both extracts did not affect the purinergic neurotransmission or induce muscle damages. Our results demonstrate that O. sambaquiensis and C. quadrumanus extracts significantly interfere with the noradrenergic neurotransmission without altering purinergic response or smooth muscle structure on rat vas deferens. Such results bring to light the pharmacological potential of O. sambaquiensis and C. quadrumanus molecules for therapeutics focusing on noradrenergic neurotransmission.


Subject(s)
Hydrozoa , Scyphozoa , Animals , Electric Stimulation , Male , Muscle Contraction , Nematocyst , Norepinephrine , Plant Extracts , Rats , Sympathetic Nervous System
19.
Exp Neurol ; 332: 113390, 2020 10.
Article in English | MEDLINE | ID: mdl-32598929

ABSTRACT

Because environmental elements modify chronic pain development and endogenous mechanisms of pain control are still a great therapeutic source, we investigated the effects of an early exposure to environmental enrichment (EE) in a translational model of neuropathic pain. Young male rats born and bred in an enriched environment, which did not count on running wheel, underwent chronic constriction injury (CCI) of sciatic nerve. EE abolished neuropathic pain behavior 14 days after CCI. Opioid receptors' antagonism reversed EE-analgesic effect. ß-endorphin and met-enkephalin serum levels were increased only in EE-CCI group. Blockade of glucocorticoid receptors did not alter EE-analgesic effect, although corticosterone circulating levels were increased in EE animals. In the spinal cord, EE controlled CCI-induced serotonin increase. In DRG, EE blunted the expression of ATF-3 after CCI. Surprisingly, EE-CCI group showed a remarkable preservation of sciatic nerve fibers compared to NE-CCI group. This work demonstrated global effects induced by an EE protocol that explain, in part, the protective role of EE upon chronic noxious stimulation, reinforcing the importance of endogenous mechanisms in the prevention of chronic pain development.


Subject(s)
Environment , Neuralgia/prevention & control , Peripheral Nerve Injuries/complications , Sciatic Nerve/injuries , Animals , Cell Survival , Constriction, Pathologic , Endorphins/blood , Enkephalins/blood , Hyperalgesia/pathology , Male , Nerve Fibers/pathology , Neuralgia/etiology , Neuralgia/pathology , Peripheral Nerve Injuries/pathology , Rats , Rats, Wistar , Receptors, Glucocorticoid/metabolism , Sciatic Nerve/pathology , Spinal Cord/metabolism , Weight-Bearing
20.
Neurosci Lett ; 721: 134765, 2020 03 16.
Article in English | MEDLINE | ID: mdl-32004656

ABSTRACT

Previous studies suggested the pharmacological potential of rat hemopressin (PVNFKFLSH) and its shorter synthetic peptide NFKF, to protect from pilocarpine-induced seizures in mice. Orally administered NFKF was shown to be hundred times more potent than cannabidiol in delaying the first seizure induced by pilocarpine in mice. Here, using an experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis we have shown that C57BL/6 J mice orally administrated with NFKF (500 µg/kg) presented better EAE clinical scores and improved locomotor activity compared to saline administrated control mice. NFKF blocked the production of IL-1beta and IL-6, and has high scores binding cannabinoid type 2 receptors. Therefore, NFKF is an exciting new possibility to neurodegenerative diseases therapeutics.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Hemoglobins/therapeutic use , Neuroprotective Agents/therapeutic use , Peptide Fragments/therapeutic use , Animals , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Hemoglobins/chemistry , Hemoglobins/pharmacology , Mice , Mice, Inbred C57BL , Molecular Docking Simulation/methods , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL