Next Article in Journal
Microneedle-Mediated Transdermal Delivery of Genetic Materials, Stem Cells, and Secretome: An Update and Progression
Previous Article in Journal
Physiologically Based Pharmacokinetic Modeling in Neonates: Current Status and Future Perspectives
Previous Article in Special Issue
Novel Drug Targets and Emerging Pharmacotherapies in Neuropathic Pain
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Unveiling the Pain Relief Potential: Harnessing Analgesic Peptides from Animal Venoms

by
Ana Flávia Marques Pereira
1,†,
Joeliton S. Cavalcante
2,†,
Davi Gomes Angstmam
2,
Cayo Almeida
3,
Gean S. Soares
4,
Manuela B. Pucca
5 and
Rui Seabra Ferreira Junior
1,2,6,*
1
Center for the Study of Venoms and Venomous Animals (CEVAP), São Paulo State University (UNESP—Univ Estadual Paulista), Botucatu 01419-901, SP, Brazil
2
Graduate Program in Tropical Diseases, Botucatu Medical School (FMB), São Paulo State University (UNESP—Univ Estadual Paulista), Botucatu 01419-901, SP, Brazil
3
Center of Mathematics, Computing Sciences and Cognition, Federal University of ABC, Santo André 09280-560, SP, Brazil
4
Delphina Rinaldi Abdel Azil Hospital and Emergency Room (HPSDRAA), Manaus 69093-415, AM, Brazil
5
Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University, Araraquara 14801-320, SP, Brazil
6
Center for Translational Science and Development of Biopharmaceuticals FAPESP/CEVAP, São Paulo State University (UNESP—Univ Estadual Paulista), Botucatu 01419-901, SP, Brazil
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceutics 2023, 15(12), 2766; https://doi.org/10.3390/pharmaceutics15122766
Submission received: 19 September 2023 / Revised: 8 November 2023 / Accepted: 5 December 2023 / Published: 13 December 2023

Abstract

:
The concept of pain encompasses a complex interplay of sensory and emotional experiences associated with actual or potential tissue damage. Accurately describing and localizing pain, whether acute or chronic, mild or severe, poses a challenge due to its diverse manifestations. Understanding the underlying origins and mechanisms of these pain variations is crucial for effective management and pharmacological interventions. Derived from a wide spectrum of species, including snakes, arthropods, mollusks, and vertebrates, animal venoms have emerged as abundant repositories of potential biomolecules exhibiting analgesic properties across a broad spectrum of pain models. This review focuses on highlighting the most promising venom-derived toxins investigated as potential prototypes for analgesic drugs. The discussion further encompasses research prospects, challenges in advancing analgesics, and the practical application of venom-derived toxins. As the field continues its evolution, tapping into the latent potential of these natural bioactive compounds holds the key to pioneering approaches in pain management and treatment. Therefore, animal toxins present countless possibilities for treating pain caused by different diseases. The development of new analgesic drugs from toxins is one of the directions that therapy must follow, and it seems to be moving forward by recommending the composition of multimodal therapy to combat pain.

1. Introduction

The International Association for the Study of Pain provides a definition of pain as “an unpleasant sensory and emotional experience associated with, or similar to that associated with, actual or potential tissue damage” [1]. Pain can be either acute or chronic, mild or severe, constant or intermittent, throbbing or steady, and can be categorized as somatic or visceral. However, describing pain accurately, along with determining its precise location, is intricate, as it might be confined to a particular spot or encompass a widespread area. Comprehending the origins and mechanisms responsible for these pain variations is imperative for implementing proper management and pharmacological interventions.
In this context, animal venoms have been demonstrated over the years to be potential sources of biomolecules that can be used as analgesic agents across a diverse range of pain models [2]. A vast array of animals, including annelids, cnidarians, echinoderms, mollusks, arthropods, and vertebrates, yield venoms, leading to a significant variability in their composition. While the potential harm posed by venomous animals is known, they have also been harnessed as therapeutic agents to treat a wide variety of inflammatory and infectious diseases [3,4,5].
This review aims to highlight the most promising venom-derived toxins that have been investigated as potential prototypes for analgesic drugs. Furthermore, it seeks to encourage scientists to explore untapped taxa for prospective analgesics and delve into the mechanisms of action of already identified molecules, some of which have been synthesized. The discussion extends to research perspectives and the obstacles to using toxins to obtain new analgesic molecules.

2. Pain Pathophysiology

Pain is a complex experience involving an actual or potential stimulus of something harmful and the physiological and emotional reactions to these events. Pain remains subjective, with each individual presenting the sensation uniquely through personal experiences. The individual then imparts unpleasant experiences across three distinct dimensions: (i) emotion, (ii) imagination, and (iii) sensation [6].
The noxious stimulus is detected by specialized peripheral sensory neurons known as nociceptors. These nociceptors have a dual projection—one extends as a peripheral axon to the skin and other organs, while the other projects toward the central nervous system (CNS) [7,8] (Figure 1). Following an injury, chemical mediators synthesized within the tissue undergo conversion into an electrical signal by transient receptor potential generator channels (TRP channels) and purinergic channels. Upon transduction, these neuronal events are transmitted by sensory fibers that project to Rexed’s lamina. Subsequently, from the spinal cord, the information travels to the brainstem and reaches the cerebral cortex, culminating in the perception of pain [9].
Physiological pain presents a protective purpose, yet it transforms into a pathological phenomenon in several instances (e.g., neuropathy and inflammation). Under these conditions, individuals and animals exhibit sensitivity to painful stimuli (hyperalgesia) [10], as well as responses to mechanical stimuli (mechanical allodynia) [11] and thermal (thermal allodynia) [12]. The pain can be categorized into nociceptive, neuropathic, and inflammatory when we considering pathological circumstances. In cases of physiological pain, the perception of it arises from nociception, the process through which noxious stimulation is relayed via the peripheral and central nervous systems.

2.1. Nociceptive Pain

Nociception is the response of the sensory nervous system to potentially harmful chemical, mechanical, and thermal stimuli, as well as actual or potentially harmful damage, triggered by the activation of nociceptors. Aδ and C fibers are the two types of nociceptors that have specialized free nerve endings and are widely located in the skin, muscles, joint capsules, bones, and some important internal organs [9].

2.2. Neuropathic Pain

Neuropathic pain is a type of chronic pain caused by damage to any level of the nervous system, either in peripheral regions, injuring peripheral fibers, or in regions of the central nervous system, such as the spinal cord and/or the brain [13]. Such diseases and/or injuries disrupt the normal activity of ion channels (sodium, calcium, and potassium), inhibitory interneurons, and descending modulatory control systems that modify the conduction of sensory signals to the spinal cord and the brain [14,15].
Common conditions associated with peripheral neuropathic pain include painful diabetic polyneuropathy [16], postherpetic neuralgia [17], trigeminal neuralgia [18], HIV-associated neuropathic pain [19], cancer-related pain [20], nerve compression (e.g., carpal tunnel syndrome herniated disc) [21], post-surgical pain (e.g., following mastectomy [22], herniorrhaphy [23], phantom limb pain [24], and others), and neuropathy from toxic exposure (e.g., antiretroviral agents, chemotherapeutics, and antituberculosis drugs) [25]. On the other hand, post-traumatic spinal cord pain, pain related to multiple sclerosis, post-stroke pain, transverse myelitis, post-radiation myelopathy, and HIV myelopathy are examples of central neuropathies.

2.3. Inflammatory Pain

Inflammatory pain arises as an inflammatory cascade is triggered, which then sensitizes peripheral nerve endings, with the aim of eliminating necrotic cells and initiating the process of tissue repair [26,27,28]. This type of pain can be classified as (i) acute inflammatory pain, which is usually intense and occurs in a short period of time; (ii) chronic inflammatory pain, which manifests with diverse levels of pain and persists beyond the period of healing [29]. After injury or damage, nociceptor-modulated peripheral sensitization occurs, causing primary hyperalgesia (exaggerated sensitivity to pain), or primary allodynia (sensation of pain when the stimulus is not normally painful). Chemical mediators such as histamine, bradykinin, acids, and serotonin are released and have the capacity to activate these nociceptors, making them depolarized or sensitized. Consequently, nociceptor terminals become more receptive to the same concentration of chemical mediators, enhancing their sensitivity [30,31].
Central sensitization is the amplification of pain by central nervous system mechanisms, which occurs independently of peripheral injury or inflammation. Repetitive and usually high-intensity synaptic transmission activates intracellular signal transduction cascades in dorsal horn neurons that enhance response to subsequent stimuli [32,33]. Several neurotransmitters participate in the perception of pain. This encompasses major types of both inflammatory and non-inflammatory mediators, second messenger production, their interaction with different channel-linked receptors, and the ensuing pharmacological effects at the terminals of both pre- and post-synaptic neurons (Figure 2). AMPA, NMDA, and metabotropic glutamate receptors, as well as the substance P (neurokinin) receptor NK1, and the BDNF (neurotrophin) receptor TrkB expressed by dorsal horn neurons, are involved in the induction of central sensitization [6]. In pre-synaptic sites, the activation of cytokine receptors and chemokine receptors results in phosphorylation and the activation of MAPK family members (ERK and p38), leading to the release of glutamate from synaptic vesicles via the activation of ion channels [34]. On the other hand, at post-synaptic sites there is an increase in neurotransmitters’ release (e.g., glutamate). When occur this release, prompts the activation of the MAPK family, which, in turn, can lead to central sensitization by upregulating NMDA (NMDAR) and AMPA (AMPAR) receptors downregulating potassium channels [9].

3. Animal Venoms: Composition and Effects

Due to advances in the development of analytical tools, numerous venom-producing animal species, including snakes [35], scorpions [36], spiders [37], bees [38,39,40], frogs [41], and mollusks [42,43,44], among other animals, have their venoms studied in terms of the composition and identification of toxins and peptides, providing an overview of the composition of these venoms. This allowed us to understand that venoms are multifunctional and complex systems, comprising proteins with and without enzymatic activities, peptides, and amino acids, among other components.
Snake venoms are true mutable substances, composed of almost unique formulations [45,46,47,48,49], featuring in its composition mainly proteins, peptides, and their respective isoforms. These molecules are distributed in dominant families: secreted A2 phospholipases (PLA2s), snake venom metalloproteinases (SVMPs), snake venom serine proteases (SVSPs), and three-finger peptides (3FTXs). Also we can cite secondary secretory proteins, kunitz peptides, L-amino acid oxidase (LAAO), the cysteine-rich secretory protein (CRiSP), C-type lectins and C-type lectin-like (CTL/SNACLEC), disintegrin (DISI) and natriuretic peptides (NPs), including vasoactive peptides, bradykinin potentiating peptides, and inhibitory peptides [35].
Some venoms are able to induce pain [6,42], edema, local and systemic inflammatory process [50,51,52], oxidative stress [53], the functional destabilization of the coagulation system [54,55], acute kidney injury [56], rhabdomyolysis, and necrosis. In addition, other snake venoms can trigger a neurotoxic syndrome: flaccid paralysis (bilateral ptosis and ophthalmoplegia) and flaccid neuromuscular paralysis (descending), which can be aggravated by involving the bulbar blockade (mouth and throat muscles responsible for speech and swallowing) and respiratory muscles [57].
Scorpion venoms are characterized by various peptides that are responsible for triggering physiopathological manifestations upon envenomation. Following envenomation, these peptides interact with ion channels, while the enzymes present in the venom facilitate the biodistribution of venom components [58]. The components of scorpion venoms and their encoding transcripts are classified into eight main categories: (i) Na+ channel toxins, (ii) K+ channel toxins, (iii) Ca2+ channel toxins, (iv) Cl channel toxins, (v) TRP channel toxins, (vi) enzymes, (vii) protease inhibitors, and (viii) defense peptides [36]. During envenomation by Buthidae scorpions, clinical manifestations range from localized signs, systemic manifestations, and vital insufficiency to dangerous cardiorespiratory symptoms, often including cardiogenic shock, pulmonary edema, or severe neurological impairment (coma and/or convulsions) [59].
The use of venom is also a success story for spider predation purposes due to its effectiveness against a wide variety of prey and in defense. Although the effect of spider venom toxins on humans is overestimated, bites that result in severe envenomation are limited to a restricted number of spider taxa and include mygalomorph Australian funnel-web spiders (Atrax and Hadronyche, Atracidae), the araneomorph recluse spiders (Loxosceles, Si-cariidae), widow spiders (Latrodectus sp., Theridiidae), and wandering spiders (Phoneutria, Ctenidae). The ability to induce pathophysiological manifestations in humans also directly influences research on the venom. Therefore, the composition of spider venom was for a long time largely restricted to specific taxa that have public health relevance. In general, spider venoms are divided into four groups: (i) small-molecular-mass compounds (SMMSs) which include ions, organic acids, nucleotides, nucleosides, amino acids, amines, and polyamines; (ii) antimicrobial peptides (cytolytic or cationic) that are found in only a few families of spiders; (iii) peptide neurotoxins that modulate a wide variety of channels and receptors present in the membranes of excitable cells (for example, nerves and muscles); and (iv) proteins and enzymes, which include cysteine-rich secretory proteins (CRISPs), hyaluronidases, collagenases, and phospholipases. These, in turn, act on the extracellular matrix or cell membrane, facilitating the movement of toxins into the prey as so-called dissemination factors [37].
Bee venom is a complex mixture of compounds, which include proteins, peptides, amino acids, phospholipids, sugars, biogenic amines, volatile compounds, pheromones, and a large amount of water (>80%). We can also find present in the venom, adrenaline, dopamine, histamine, hyaluronidase, noradrenaline, phospholipases A 2 (PLA2s), phospholipases B (PLBs), serotonin, apamine, melittin, and mast cell degranulator peptide (MCD) [38,39,40]. Honeybee envenomation may cause anaphylactic shock, and in multiple stings, entire organ systems can be affected, cause cardiovascular diseases, pulmonary dysfunction, renal failure, hepatic damage, and hematologic diseases [60].
Although there is no apparent clinical relevance to human health, frog poisonings are commonly reported in domestic animals, highlighting their veterinary importance. The venoms secreted by the parotid glands of frogs correspond to a mixture of biologically active compounds, including dopamine, epinephrine, norepinephrine, serotonin, bufotenin, bufagenins, bufotoxins, and indolealkylamines, which are rapidly absorbed by the mucous membranes (including the gastric mucosa, in the cases of ingestion). Upon entering the systemic circulation, the greatest effects are seen in the heart, the peripheral vasculature, and the nervous system [41,61]
The venomous mollusks from the superfamily Conoidea have venoms with 100 to 200 components in their composition. There are small peptides conotoxins of 5–50 amino acids that have high biological activity. These peptides are classified according to number of cysteine residues and pharmacological targets. The most extensively researched conotoxins include α, ω, κ, μ, and δ toxins, that acts inhibiting nicotinic acetylcholine receptors, voltage-gated calcium channels, voltage-gated potassium channels, voltage-gated sodium channels, and voltage-gated sodium channels, respectively. The Conus species envenomation may be fatal, causing immobilization and the inhibition of neuromuscular transmission, leading to cardiac and neurological disorders [42,43,44].
Animal venoms have shown a high potential for the development of products, some of which are already used clinically [62]. This is due to the range of molecules with different biological functions, which make animal venoms a true source for the mining of new compounds with potential, both for use in basic research and for the development of new drugs (antitumor, antimicrobial, antiviral, and anti-inflammatory drugs, among others). In contrast, peptide toxins are generally found in lesser abundance; however, their diversity as well as ease of synthesis have attracted substantial attention. Despite their diversity, venom peptides appear to have evolved from a relatively small number of structural structures that are particularly suited to addressing the crucial issues of potency and stability, which makes them a unique source of linkages and structural models important to build new therapeutic agents.

4. Toxins Targeting Pain: Discovering Potential Analgesics

During pain signaling, nociceptors and post-synaptic neurons are activated or inhibited by neurotransmitters binding to many channels and transporters that contribute to the release of pain-related neurotransmitters and/or ion fluxes relevant to stimulus conduction. Pain can be modulated by venom-derived toxins, such as by selective toxins that bind to receptors responsible for modulating the detection, conduction, and propagation of painful impulses, enabling the use of these toxins in the design and development of novel analgesic drugs. Despite the variety of pharmacological targets, ion channels have drawn attention. Voltage-gated sodium channels (VGSCs) and transient receptor potential (TRP) channels are crucial therapeutic targets in pain management, which form the critical components of the nociceptive sensory pathway.
The initial investigations reporting the analgesic activities of animal venoms primarily focused on snake venoms, with some studies mentioning bee venom. From 1970 to 2004, there was an exponential increase in the frequency of published articles that investigated venoms and toxins in relation to nociception. After 2004, the publication trend diverged from the previously observed pattern of pain-related articles, reaching a plateau that resulted in a linear progression in the field of venom and toxin research [63]. This change in trajectory could potentially be attributed to the discovery of novel venoms, along with the development of methodologies for evaluating their effectiveness. Therefore, we conducted a contemporary analysis, meticulously selecting bioprospecting investigations involving analgesic peptides examined in animal models.

4.1. Snake Analgesic Peptides

Several toxins purified and synthesized from snake venom have biotechnological potential for the treatment of various human diseases [64,65,66,67]. As far as the search for new analgesic drugs is concerned, snake venoms have been little explored, perhaps due to their widely reported potential for inducing algesia in human envenomations. However, crotoxin (CTX) isolated from Crotalus spp. venoms, Najanalgesin and Cobra neurotoxin from Naja naja atra venom, Mambalgin-1 and Mambalgin-3 from Dendroaspis polylepis polylepis venom [68], µ-EPTX-Na1 from D. polylepis polylepis venom, and µ-EPTX-Na1 from Naja attracts [69] have demonstrated a variety of analgesic activities.
CTX induces analgesia via formyl peptide, α2-adrenergic, and muscarinic receptors in the acute. Also we can observe in chronic phases of hypernociception induced via partial sciatic nerve ligation [70]. It is dependent on formyl peptide, lipoxygenase, and muscarinic receptors, as shown in MOG 35-55-induced experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis [71]. The previously outlined crotalfine, described in earlier years, has recently demonstrated its ability to intervene in mitigating the desensitization process of TRPA1 receptors [72]. This result in the induction of analgesia in pain protocols, specifically within the contexts of cold hypersensitivity through ciguatoxin-induced expression and and also mechanical hypersensitivity provoked by bradykinin- and zymosan-induced expression [73]. Najanalgesin and cobra neurotoxin induce analgesia through adenosine receptor (A1 and A2A) pathway activation in acute pain models induced by hot plate and spinal cord injury [74] and the c-Jun N-terminal kinase (JNK) inhibitor in neuropathic pain induced by spinal nerve ligation [75]. However, the induction of analgesia promoted by µ-EPTX-Na1 involves the Nav1.8 channel inhibitor [69], while mabalgin 1 and 3 present a common analgesia mechanism via acid-sensing ion channel (ASIC) inhibitors, as evidenced by the motor behavior tests such as the accelerated rotarod test and the grip strength test. This phenomenon can be observed in pain and inflammation models induced by carrageenan and thermal or mechanical von Frey tests. Furthermore, the isolated Cobratoxin from the venom of Naja naja kaouthia has exhibited analgesic effects in acute pain models targeting the α7 subunit of nicotinic acetylcholine receptors when administered intrathecally, thereby eliciting an antinociceptive response in mice [76] (Table 1).

4.2. Scorpion Analgesic Peptides

Scorpionism is a major public health problem in subtropical areas worldwide even though less than 25 species are considered dangerous to humans. The “Old World” (Africa) species of medical interest belong to the genera Andoctonus, Buthus, Hottentota, and Leiurus, while the “New World” (America) species belong to the Centruroides and Tityus genera, belonging to the Buthid family. One of the most prominent clinical signs following a scorpion sting is pain, which is mainly caused by neurotoxins targeting ion channels. Remarkably, the same scorpion venoms able to induce pain mediated by several mechanisms also exhibited analgesic activities, thereby establishing the groundwork for a potential pain-relieving agent.
Different toxins from the scorpion Buthus martensii are promising for the treatment of pain in a xenograft tumor mouse model [77,78]. These toxins can be important candidates also to use in acute thermal pain model induced by a hot plate, in acute inflammation model induced by formalin [79,80,81,82], in a mouse-twisting pain model, mechanical allodynia, in a nociception model induced by a shutter-controlled lamp, and in an inflammation model induced by formalin and acetic acid writhing [81]. Furthermore, TsNTxP isolated from Tityus serrulatus venom proved to be a good candidate in acute nociception induced by a water bath and a neuropathic pain model induced by the constriction of a sciatic nerve (CCI model) [83], while Leptucin from Hemiscorpius lepturus venom showed analgesic potential against an acute thermal pain model induced by a hot plate and a nociception model using the tail-flick test [84].
Peptides derived from scorpion venoms exhibit a wide variety of analgesic mechanisms: (i) ion channel inhibitors; (ii) MCF-7 and MDA-MB-231 migration inhibitors; (iii) MAPK inhibitors; (iv) arginine residue at position 58; and (v) CHis6-rAGAP NHis6-rAGAP. The various pharmacological mechanisms of scorpion venoms have likely drawn considerable attention to the bioprospecting due peptides present in this venom, while the venom of other species/genera remains underexplored and even unknown (Table 2).

4.3. Spider Analgesic Peptides

Spiders (order Araneae) currently comprise 49,483 species, almost all of which have the capacity to produce venom (WSC, 2021). However, in recent years, the exploration of spider venoms in search of prototype peptides for the design of new analgesic drugs has been restricted to a small number of genera. There is extensive research involving Phoneutria nigriventer peptides, some isolated from the venom, while others synthesized and/or obtained recombinantly. The venom of this spider species has represented an extensive shelf of compounds with potential for the development of new analgesic drugs in different pain models: inflammatory pain; cold/heat/mechanical hyperalgesia and allodynia; post-operative pain; hyperalgesia; neuropathic pain; cancer-related pain; mechanical pain; fibromyalgia; and others [87,88,89,90,91] (Table 3).
Peptides from P. nigriventer exhibit a wide variety of analgesic mechanisms, which include (i) a reduction in glutamate levels in the cerebrospinal fluid [92]; (ii) an effect on peripheral opioid and cannabinoid systems [93,94]; (iii) the activation of the NO-cGMP pathway -KATP [95]; (iv) the inhibition of spinal AChE resulting in the activation of muscarinic and nicotinic receptors [96]; (v) the selective antagonism of TRPA1 channels [92]; (vi) the reversible inhibition of voltage-gated calcium channels (VGCC) [97]; and (vii) the antagonism of the CXCR4 receptor [98]. There is a great diversity of pharmacological mechanisms in peptides of Phoneutria nigriventer venom, but the venom of other species/genera remains underexplored and even unknown.
The potential for the treatment of pain in underexplored spider venoms is mainly due to the inhibition of ion channels that control the transit of neurotransmitters. Furthermore, an interesting potential was reported by Deuis et al. [99] for μ-theraphotoxin-Pn3a isolated from the venom of Pamphobeteus nigricolor. The intraperitoneal injection of this peptide induces analgesia by inhibiting Nav1.7 channels and exhibited synergy with opioids in a model of peripheral pain induced by the injection of α-scorpion toxin OD1. Other peptides isolated from Ceratogyrus sp., Davus sp., Haplopelma lividum, Heteropoda venatoria, Thrixopelma pruriens, and Grammostola porteni also present potent candidates for analgesic drugs through the modulation of ion channels [79,87,88,90,91,96,99,100,101,102,103,104].
Table 3. Spider-derived toxins able to inhibit pain.
Table 3. Spider-derived toxins able to inhibit pain.
Toxin/
Molecule
Species ProductionAdministrationMechanism of AnalgesiaModelRef.
PhTx3-4Phoneutria nigriventerPurified from venomIntrathecal (i.t.) injectionReduction in glutamate levels in the cerebrospinal fluidPersistent inflammatory pain and post-operative (plantar incision) nociception in mice[92]
PnPP-19Phoneutria nigriventerSynthesizedSubcutaneous (s.c.) injectionBoth opioid and cannabinoid peripherical systemsHyperalgesia induced by the administration of PGE2[94]
Tx3-5Phoneutria nigriventerPurified from venomIntrathecal (i.t.) injection-Postoperative (plantar incision), neuropathic (partial sciatic nerve ligation), and cancer-related pain (inoculation with melanoma cells) in animals[105]
δ-CNTX-Pn1aPhoneutria nigriventerPurified from venomIntrathecal (i.t.) injectionOpioid and
cannabinoid systems
Hyperalgesia induced by the CCI model and hyperalgesia induced by the administration of PGE2[93]
PnPP-19Phoneutria nigriventerSynthesizedSubcutaneous (s.c.) injectionActivation of NO-cGMP-KATP pathwayHyperalgesia induced by the administration of PGE2[95]
PhKvPhoneutria nigriventerPurified from a PhTx3 fractionIntrathecal (i.t.) injectionInhibition of spinal AChE resulting in the activation of the muscarinic and nicotinic receptorsChronic constriction injury model and after intraplantar injection of capsaicin[97]
Phα1βPhoneutria nigriventerPurified from venomIntrathecal (i.t.) or intraplantarly (i.pl.) injectionsSelective antagonist of TRPA1 channelsNocifensive responses evoked by reactive TRPA1 channel agonist, mechanical and cold hyperalgesia, neuropathic pain induced by the chemotherapeutic agent bortezomib[106]
Pha1βPhoneutria nigriventerPurified from venomIntrathecal (i.t.) injectionReversibly inhibits the voltage-gated calcium channels (VGCC)Nociception that was triggered by capsaicin, CCI model, and hyperalgesia was induced in the melanoma cancer pain model[97]
Pha1βPhoneutria nigriventerRecombinantIntrathecal (i.t.) injectionReversibly inhibits the voltage-gated calcium channels (VGCC)Nociception that was triggered by capsaicin, CCI model, and hyperalgesia was induced in the melanoma cancer pain model[97]
PnTx4(5-5)Phoneutria nigriventerPurified from venomIntraplantarly (i.pl.) and subcutaneous (s.c.) injectionThe antinociceptive effect of PnTx4(5-5) can also be related to the glutamatergic systemHyperalgesia induced by PGE2, carrageenan, and L-glutamate (L-Glu)[107]
Phα1βPhoneutria nigriventerRecombinantIntravenous (i.v.) injection-Pain was induced by the CCI model and paclitaxel-induced acute and chronic pain[108]
CTK
01512-2
Phoneutria nigriventerRecombinantIntrathecal (i.t.) injectionReversibly, and not specifically a block of Cav 2.2 that affects the intracellular Ca2+ influx and the glutamate releaseMechanical and thermal hyperalgesia and cold allodynia[109]
Phα1βPhoneutria nigriventerPurified from venomIntrathecal (i.t.) injectionBlocker voltage-dependent calcium channels and the antagonism with the receptor CXCR4Diabetic neuropathic pain[98]
PnAn13Phoneutria nigriventerSynthetic peptideIntrathecal (i.t.) injectionOpioid and
cannabinoid systems
Hyperalgesia induced by PGE2[110]
Phα1βPhoneutria nigriventer-Intrathecal (i.t.) injectionBlocker TRPA1 and Cav2.2 receptorsPostoperative (plantar incision)[111]
Phα1βPhoneutria nigriventerRecombinantIntravenous (i.v.) injectionHigh-voltage calcium channel inhibitors (HVCCs) and cationic channel antagonists of the potential transient receptor (TRPA1)Hyperalgesia and mechanical allodynia induced by reserpine (fibromyalgia)[112]
Cd1aCeratogyrus darlingiChemical synthesisIntraplantar (i.pl.) injectionNav/Cav channel inhibitorPeripheral
pain by NaV channels induced with α-scorpion toxin OD1
[87]
JZTX-XChilobrachys jingzhaoChemical synthesisIntraplantar (i.pl.) injectionKv4 channel inhibitorMechanical hypersensitivity[100]
μ-TRTX-Ca2aCyriopagopus albostriatusPurified from venom Intrathecal (i.t.) and intraplantar (i.pl.) injectionNa v 1.7 channel inhibitorAcute inflammation models induced by formalin and acetic acid and acute thermal pain models induced by hot plate[101]
µ-TRTX-Ca1a Cyriopagopus albostriatusPurified from venomIntraperitoneal (i.p.) injectionhNa v 1.7 channel inhibitorPain formalin-induced paw licking, hot plate test, and acetic acid-induced writhing [102]
CyrTx-1aCyriopagopus schioedteiPurified from venomIntraperitoneal (i.p.) injectionhNa V 1.7 channel inhibitorThermal hyperalgesia[103]
Df1a-NH2 Davus fasciatusChemical synthesisIntraplantar (i.pl.) injectionNaV and CaV3 channel inhibitorPeripheral
pain by NaV channels induced with α-scorpion toxin OD1
[88]
Df1a-OHDavus fasciatusChemical synthesisIntraplantar (i.pl.) injectionNaV and CaV3 channel inhibitorPeripheral
pain by NaV channels induced with α-scorpion toxin OD1
[88]
GpTx-1 and GpTx-1-71Grammostola porteniChemical synthesisIntrathecal (i.t.) and Intracerebroventricular (i.c.v.) injectionNav channel inhibitorTail-flick test, carrageenan- or complete Freund’s adjuvant (CFA)-induced inflammatory pain model, neuropathic pain model, mechanical allodynia, thermal hyperalgesia, writhing test, formalin test, tolerance evaluation, rotarod test, open field test, gastrointestinal transit test
[104]
HpTx3Heteropoda venatoriaPurified from venomIntramuscular (im.i) and intraperitoneal (i.p.) injectionsNav1.7 channel inhibitorAcute inflammation models induced by formalin and acetic acid, chronic inflammation pain models induced by complete Freund’s adjuvant, acute thermal pain models induced by hot plate, and chronic neuropathic pain models induced by spared nerve injury[90]
μ-theraphotoxin-Pn3a Pamphobeteus nigricolorPurified from venomIntraperitoneal (i.p.) injection Nav1.7 channel inhibitor and synergy with opioidsPeripheral
pain by NaV channels induced with α-scorpion toxin OD1
[99]
ProTx-III Thrixopelma pruriensRecombinantIntraplantar (i.pl.) injectionNav1.7 channel inhibitorPeripheral
pain by NaV channels induced with α-scorpion toxin OD1
[91]
ProTx-II (β/ω-terafotoxina-Tp2a)Thrixopelma pruriens-Intracerebroventricular (i.cv) injectionNav1.7 channel inhibitorAcute thermal pain model induced by hot plate and mechanical allodynia[96]

4.4. Frogs Analgesic Peptides

Little is known about anuran venoms, which is partly due to the low number of poisonings with these animals. Anurans have granular glands that produce venoms composed of peptides, biogenic amines, steroids, and alkaloids, which induce cardiotoxic, neurotoxic, myotoxic, anesthetic, antimicrobial, hemolytic, healing, antidiabetic, and other effects [61,113]. Although the potential of the venoms of these animals is recognized, only the peptide bufalin, purified from the venom of Bufo gargarizans, was studied as an analgesic agent in an animal model [114]. Intraperitoneally, bufaline was able to inhibit the pain caused via the injection of formalin and carrageenan and presented pharmacological potential against mechanical and thermal allodynia via the inhibition of the activities of VGSCs [114] (Table 4).
Table 4. Frog-derived toxins able to inhibit pain.
Table 4. Frog-derived toxins able to inhibit pain.
Toxin/
Molecule
Species ProductionAdministrationMechanism of AnalgesiaModelRef.
BufalinBufo gargarizansPurified from venomIntraperitoneal (i.p.) injectionVGSC activity inhibitorPain formalin-induced paw licking, pain carrageenan-induced thermal and mechanical hyperalgesia[114]

4.5. Bee Analgesic Peptides

The Apis mellifera venom has peptides, bioactive compounds, and proteins in its composition. The main components are melittin (most abundant representing 40–60% of the dry weight of the venom), phospholipase A2 (bvPLA2), and apamin (a peptide). Some properties of this venom have already been reported in the literature, such as its anticancer, antinociceptive, and anti-inflammatory properties [115]. Bioprospecting studies has been carried out with the raw venom of A. mellifera bees to show the ability to induce analgesia in pain caused by spinal cord injury (SCI)-induced allodynia; thermal hyperalgesia [116]; oxaliplatin-induced mechanical allodynia [117]; complex regional pain syndrome type 1 (CRPS-I)-induced mechanical allodynia [118]; oxaliplatin-induced neuropathic pain, cold and mechanical allodynia [119]; vincristine-induced cold and mechanical hypersensitivity [120]; scalding-burn-model-induced mechanical allodynia [121]; and paclitaxel-induced cold and mechanical allodynia [120,122] (Table 5).
A. mellifera venom and its toxins cause analgesia through (i) effects on α-1 adrenergic and α-2 adrenergic receptors [116,120,122,123]; (ii) serotonergic (5-HT1/5-HT2 and 5-HT3) and opioid receptors [120]; (iii) the inhibition of COX expression [118,124]; (iv) the suppression of glial cells [121]; (v) the suppression of NK-1 receptor expression in mice with complex regional pain syndrome type I (CRPS I) [118]; (vi) an increase in the A-fiber action potential threshold of DGR neurons [119]; and (vii) the inhibition of the expression of substance P in the central and peripheral nervous system [121].
Table 5. Bee venom able to inhibit pain.
Table 5. Bee venom able to inhibit pain.
Toxin/
Molecule
Species ProductionAdministrationMechanism of AnalgesiaModelRef.
Bee venomApis melliferaSigma-Aldrich®Subcutaneous
(i.s.) injection
Suppression of glial cell activation ipsilateral, dorsal spinal cordSpinal cord injury (SCI)-induced allodynia and thermal hyperalgesia[116]
Bee venomApis melliferaSigma-Aldrich®Subcutaneous (i.s.) injectionα-2 adrenergic receptors
activation
Oxaliplatin-induced mechanical allodynia[117]
Bee venomApis mellifera-Subcutaneous (i.s.) injectionDecrease of
NK-1 receptor expression in dorsal root ganglia (DRG)
Complex regional pain syndrome type-1 (CRPS-I) induced mechanical allodynia[118]
Bee venomApis melliferaJayeonsaeng TJ®Subcutaneous (i.s.) injectionIncrease of the action potential threshold in A-fiber DRG neuronsOxaliplatin-induced neuropathic pain, cold and mechanical allodynia[119]
Bee venomApis melliferaJayeonsaeng TJ®Subcutaneous (i.s.) injectionα-2 adrenergic receptors
activation with the involvement of noradrenergic nuclei
of the locus coeruleus (LC)
Vincristine-induced cold and mechanical hypersensitivity[120]
Bee venomApis melliferaApis Injeel®Intraperitoneal (i.p.)
Injection
Inhibitory activity of the COX pathwayComplete Freund’s adjuvant-induced arthritic rats[124]
Bee venomApis melliferaSigma-Aldrich®Subcutaneous (i.s.) injectionInhibitory effect on the expression of substance P in the peripheral and central nervous systemsScalding-burn-model-induced mechanical allodynia[121]
Bee venom
+ Morphine
Bee venom from Apis melliferaSigma-Aldrich®Intrathecal (i.t.) injectionSpinal opioidergic and 5-HT3 receptors modulate the analgesiaOxaliplatin-induced neuropathic pain, cold and mechanical allodynia[125]
Bee venom
+ Venlafaxine
Bee venom from Apis melliferaJayeonsaeng TJ®Intrathecal (i.t.) injectionα-2 adrenergic receptors
activation and serotonergic receptors (5-HT1/5-HT2 and 5-HT3)
Paclitaxel-induced cold and mechanical allodynia[126]
Bee venom and melittinApis melliferaPurified from bee venomSubcutaneous (i.s.) injectionα-2 adrenergic receptor activationPaclitaxel-induced mechanical hyperalgesia[122]
MelittinApis melliferaSigma Aldrich®Intraprostatic injectionSuppression of COX-2 expressionComplete Freund’s adjuvant-induced prostatitis[127]
Melittin Apis melliferaPurified from bee venom—obtained from Sigma-Aldrich®Subcutaneous (i.s.) injectionSpinal α-1 and α-2 adrenergic receptor activationOxaliplatin-induced mechanical and cold allodynia[128]
Phospholipase A2 (bvPLA2)Apis melliferaSigma-Aldrich®Intraperitoneal (i.p.) injectionActivation of the noradrenergic system, via α2-adrenergic receptorsOxaliplatin-induced neuropathic pain—cold and mechanical allodynia[123]
bvPLA2Apis melliferaSigma-Aldrich®Intraperitoneal (i.p.) injectionSuppressing immune responses in the DRG by regulatory T cells (Tregs)Oxaliplatin-induced neuropathic pain in Treg-depleted mice[128]

4.6. Mollusk Analgesic Peptides

Conotoxins or conopeptides are small peptides presenting from 10 to 35 amino acids obtained from the venom of cone snails. They encompass a wide range of bioactive compounds, including those with analgesic potential, as is the case of the medicine Ziconotide (Prialt®), which is derived from a Conus magus conotoxin (ω-conotopeptide MVIIA) and is used for the treatment of chronic pain [129,130]. There are other marine snails with potential venoms, which are distributed in three families, namely Conoidea (containing the Conus genus), Terebridae (these are the “auger snails”), and Turridae (composed of the “turrids”), although the most studied and known peptides belong to the Conus genus [129].
Mollusk venoms have also been studied and represent a truly rich source of bioactive peptides. Among their diverse effects, their analgesic property has already been evidenced in pain models such as the rat NLP model [131] and the hot-plate model [132], as well as in rat partial sciatic nerve injury (PNL model), chronic constriction injury pain (CCI model) [133,134,135,136], chronic visceral hypersensitivity (CVH model) [137], oxaliplatin-induced neuropathic pain cold and mechanical allodynia [138], paclitaxel-induced neuropathic pain [139], neuropathy and oxaliplatin-induced neuropathic pain [140,141], post-surgical pain (PSP model), and cisplatin-induced pain [141].
Mollusk peptides act mainly via the inhibition of high-voltage-activated N-type calcium channel currents (Cav2.2) in isolated mouse dorsal root ganglia (DRG) neurons and the inhibition of G protein-coupled γ-aminobutyric acid type B receptor (GABABR)-coupled Cav2.2 channels in rat DRG neurons, which results in a reduction in the excitability of DRG neurons [134,135,141,142]. In addition, other mechanisms through which mollusk peptides induce analgesia have already been reported, such as the inhibition of TTX (tetrodotoxin)-resistant sodium currents in DRG neurons [132], the inhibition of NR2B ion channels [141], the inhibition of α9-containing nicotinic acetylcholine receptors (nAChRs) [143], and the activation of GABABRs [136]. However, the analgesia mechanism of other peptides still needs to be elucidated (Table 6).

5. Challenges in Integrating Venom-Derived Toxins for Pharmaceutical Pain Relief Solutions

When it comes to developing new biopharmaceuticals derived from venom toxins, two fundamental challenges must be addressed to effectively position these compounds as viable candidates in the pharmaceutical market. The first challenge revolves around determining the most suitable mode of administration for these compounds. How can these molecules efficiently reach their intended targets using the least invasive route?
For instance, let us examine the drug Prialt® (Ziconotide), which is primarily administered intrathecally to patients [150]. Despite the inherent advantages of this approach, such as reducing complications associated with catheters and significantly enhancing pain control efficacy, especially in advanced stages of carcinomas, this method of administration still qualifies as invasive, similar to parenteral routes like intravenous, subcutaneous, and intramuscular injections [151,152,153]. The desired goal is to adopt a route that minimizes harm to the organism and results in a lower incidence of adverse reactions. Among the considered options are oral, nasal, pulmonary, and rectal routes [151,154].
The second inherent challenge in pharmaceutical development concerning peptides and proteins revolves around the search for molecules with low mass that still possess specific biological activities. Craik et al. [155] and Lipinski [156] contend that molecules with a molecular weight below 500 Da are amenable to oral administration, while those exceeding 5000 Da forfeit this suitability due to their size. However, larger molecules demonstrate high specificity toward their target receptors, whereas smaller ones lack selectivity and often result in more side effects. It is apparent that many of the peptides currently under investigation still possess significant dimensions, which impede their ability to reach target receptors. Consequently, this affects absorption, metabolism, and distribution, ultimately making the choice of administration route more complex. These challenges underscore the complexity of developing venom-based pharmaceuticals for pain relief and emphasize the ongoing need for research and innovative approaches in this area.

6. Future Perspectives of Using Toxins as Novel Analgesics

Despite the challenges associated with the pharmaceutical utilization of peptides, the industry has made significant strides. Currently, there are over 150 peptides in clinical development, with an additional 400 to 600 peptides in various stages of preclinical trials. Moreover, there are more than 80 peptide-derived drugs already available in the market, generating a worldwide revenue exceeding USD 20 billion just in 2019. These therapeutic peptides are indicated for a wide array of conditions, including but not limited to type 1 and 2 diabetes, cancer, neuropathic pain, hypertension, osteoporosis, cardiovascular diseases, HIV, and various endocrine and metabolic disorders [155,157].
To ensure the effective action of a biomolecule at its therapeutic target, it is essential to consider the drug delivery system. This field of study focuses on methods for encapsulating proteins and peptides with the precise aim of directing them to the desired site of action [151,155,157]. Several techniques are under development, each with its own specificities, such as the use of liposomes, the spray drying technique, solid lipid nanoparticles (SLNs), double emulsions, and other approaches [158,159,160,161,162]. The objective is to safeguard the biomolecule from degradation, enabling it to reach its destination within the body effectively and thus optimizing its therapeutic capacity. The drug delivery field continues to evolve through ongoing research efforts aimed at enhancing drug delivery technologies to make treatments more efficient and safer [151,163].
Due to this promising development of drugs based on peptides, future perspectives of using toxins as novel analgesics involve strategies for reducing costs for the large-scale synthesis and purification of peptides, considering reducing costs using chemical synthesis and expression of recombinant peptides [164,165]. Currently, one of the key challenges in the bioprospecting of drugs derived from animal venoms is the acquisition of validated venom batches produced in accordance with stringent Good Production Practices (GMP) [166]. This is a costly endeavor, demanding a substantial number of animals. In situations involving animals like spiders and scorpions, the quantity of venom obtained often falls short of meeting the requirements for all stages of discovery and preclinical testing. To address these challenges, alternative methods include chemoenzymatic peptide synthesis, which relies on enzyme-mediated peptide bond formation rather than chemical reagents. This approach offers cost reductions compared to solid-phase peptide synthesis (SPPS). Additionally, automated flow peptide synthesis (AFPS) is emerging as a swifter method than traditional SPPS [157].
As we look ahead, the development of innovative peptide-derived drugs is poised to benefit from fresh perspectives on peptide design. These perspectives encompass considerations of protein–protein interactions, integration with nanotechnology-based platforms such as nanoparticles and liposomes, conjugation with proteins, the lipidation or PEGylation of peptides, and structural modifications. These approaches hold promise for optimizing drug delivery; surmounting challenges related to renal clearance; and enhancing the biological activity, stability, and solubility of peptide-based therapeutics [155,157,167].

7. Conclusions

The venoms of spiders, mollusks, scorpions, bees, frogs, and snakes offer a rich source of new pharmaceutical compounds. These venoms contain peptides that play a fundamental role in defense and predation, and they can interfere with multiple vital biological processes. Within this context, the action of numerous compounds affecting rapid synaptic transmission has provided extensive research into their pharmacological potential for pain treatment. As plausible candidates in the development of analgesics, peptides from animal venoms show selectivity for receptors associated with pain modulation in different pathological models, with different mechanisms of analgesia, which makes them valuable for proof-of-concept studies and the development of novel analgesic drugs or associated with multimodal therapies. However, for therapeutic applications, it is necessary to address a number of issues related to safety, pharmacokinetics, and delivery.

Author Contributions

Conceptualization, A.F.M.P., J.S.C. and R.S.F.J.; writing—review and editing, A.F.M.P., J.S.C., G.S.S., M.B.P., D.G.A., C.A. and R.S.F.J. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by The National Council for Scientific and Technological Development (CNPq) scholarship grant n°. 151190/2023-2 (A.F.M.P.), n°. 307184/2020-0 (M.B.P.), and n°. 301608/2022-9 (R.S.F.Jr.). We thank The São Paulo Research Foundation (FAPESP) for grant number 2023/09921-3 (A.F.M.P.), 2023/01554-1 and 2021/11936-3 Funding (R.S.F.Jr.). We also thank the Coordination of Superior Level Staff Improvement (CAPES) for scholarship grant n° 88887.674376/2022-00 (J.S.C.), n° 88887.892379/2023-00 (A.F.M.P.), n° 88887.826358/2023-00 (D.G.A.) and The São Paulo Research Foundation (FAPESP) for grant number 2022/16060-1 (J.S.C.).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Raja, S.N.; Carr, D.B.; Cohen, M.; Finnerup, N.B.; Flor, H.; Gibson, S.; Keefe, F.J.; Mogil, J.S.; Ringkamp, M.; Sluka, K.A.; et al. The Revised International Association for the Study of Pain Definition of Pain: Concepts, Challenges, and Compromises. Pain 2020, 161, 1976–1982. [Google Scholar] [CrossRef] [PubMed]
  2. Maatuf, Y.; Geron, M.; Priel, A. The Role of Toxins in the Pursuit for Novel Analgesics. Toxins 2019, 11, 131. [Google Scholar] [CrossRef] [PubMed]
  3. Utkin, Y.; Siniavin, A.; Kasheverov, I.; Tsetlin, V. Antiviral Effects of Animal Toxins: Is There a Way to Drugs? Int. J. Mol. Sci. 2022, 23, 3634. [Google Scholar] [CrossRef]
  4. Yacoub, T.; Rima, M.; Karam, M.; Sabatier, J.-M.; Fajloun, Z. Antimicrobials from Venomous Animals: An Overview. Molecules 2020, 25, 2402. [Google Scholar] [CrossRef]
  5. Sartim, M.A.; Menaldo, D.L.; Sampaio, S.V. Immunotherapeutic Potential of Crotoxin: Anti-Inflammatory and Immunosuppressive Properties. J. Venom. Anim. Toxins Incl. Trop. Dis. 2018, 24, 39. [Google Scholar] [CrossRef]
  6. dos Santos Cavalcante, J.; Júnior, F.A.; Jorge, R.J.; Almeida, C. Pain Modulated by Bothrops Snake Venoms: Mechanisms of Nociceptive Signaling and Therapeutic Perspectives. Toxicon 2021, 201, 105–114. [Google Scholar] [CrossRef]
  7. Garland, E.L. Pain Processing in the Human Nervous System. Prim. Care: Clin. Off. Pract. 2012, 39, 561–571. [Google Scholar] [CrossRef]
  8. Peirs, C.; Seal, R.P. Neural Circuits for Pain: Recent Advances and Current Views. Science (1979) 2016, 354, 578–584. [Google Scholar] [CrossRef]
  9. Yam, M.; Loh, Y.; Tan, C.; Khadijah Adam, S.; Abdul Manan, N.; Basir, R. General Pathways of Pain Sensation and the Major Neurotransmitters Involved in Pain Regulation. Int. J. Mol. Sci. 2018, 19, 2164. [Google Scholar] [CrossRef]
  10. Roeckel, L.-A.; Le Coz, G.-M.; Gavériaux-Ruff, C.; Simonin, F. Opioid-Induced Hyperalgesia: Cellular and Molecular Mechanisms. Neuroscience 2016, 338, 160–182. [Google Scholar] [CrossRef]
  11. Jensen, T.S.; Finnerup, N.B. Allodynia and Hyperalgesia in Neuropathic Pain: Clinical Manifestations and Mechanisms. Lancet Neurol. 2014, 13, 924–935. [Google Scholar] [CrossRef]
  12. Viana, F. Nociceptors: Thermal Allodynia and Thermal Pain. In Handbook of Clinical Neurology; Elsevier: Amsterdam, The Netherlands, 2018; pp. 103–119. [Google Scholar]
  13. Colloca, L.; Ludman, T.; Bouhassira, D.; Baron, R.; Dickenson, A.H.; Yarnitsky, D.; Freeman, R.; Truini, A.; Attal, N.; Finnerup, N.B.; et al. Neuropathic Pain. Nat. Rev. Dis. Primers 2017, 3, 17002. [Google Scholar] [CrossRef]
  14. Aurilio, C.; Pota, V.; Pace, M.C.; Passavanti, M.B.; Barbarisi, M. Ionic Channels and Neuropathic Pain: Phisiopatology and Applications. J. Cell Physiol. 2008, 215, 8–14. [Google Scholar] [CrossRef]
  15. Finnerup, N.B.; Kuner, R.; Jensen, T.S. Neuropathic Pain: From Mechanisms to Treatment. Physiol. Rev. 2021, 101, 259–301. [Google Scholar] [CrossRef]
  16. Kaur, S.; Pandhi, P.; Dutta, P. Painful Diabetic Neuropathy: An Update. Ann. Neurosci. 2011, 18, 168–175. [Google Scholar] [CrossRef]
  17. Mallick-Searle, T.; Snodgrass, B.; Brant, J. Postherpetic Neuralgia: Epidemiology, Pathophysiology, and Pain Management Pharmacology. J. Multidiscip. Healthc. 2016, 9, 447–454. [Google Scholar] [CrossRef]
  18. Gambeta, E.; Chichorro, J.G.; Zamponi, G.W. Trigeminal Neuralgia: An Overview from Pathophysiology to Pharmacological Treatments. Mol. Pain 2020, 16, 174480692090189. [Google Scholar] [CrossRef]
  19. Lu, H.-J.; Fu, Y.-Y.; Wei, Q.-Q.; Zhang, Z.-J. Neuroinflammation in HIV-Related Neuropathic Pain. Front. Pharmacol. 2021, 12, 653852. [Google Scholar] [CrossRef]
  20. Edwards, H.; Mulvey, M.; Bennett, M. Cancer-Related Neuropathic Pain. Cancers 2019, 11, 373. [Google Scholar] [CrossRef]
  21. Schmid, A.B.; Fundaun, J.; Tampin, B. Entrapment Neuropathies: A Contemporary Approach to Pathophysiology, Clinical Assessment, and Management. Pain Rep. 2020, 5, e829. [Google Scholar] [CrossRef]
  22. Shipton, E. Post-surgical neuropathic pain. ANZ J. Surg. 2008, 78, 548–555. [Google Scholar] [CrossRef]
  23. Niccolaï, P.; Ouchchane, L.; Libier, M.; Beouche, F.; Belon, M.; Vedrinne, J.-M.; El Drayi, B.; Vallet, L.; Ruiz, F.; Biermann, C.; et al. Persistent Neuropathic Pain after Inguinal Herniorrhaphy Depending on the Procedure (Open Mesh v. Laparoscopy): A Propensity-Matched Analysis. Can. J. Surg. 2015, 58, 114–120. [Google Scholar] [CrossRef]
  24. Subedi, B.; Grossberg, G.T. Phantom Limb Pain: Mechanisms and Treatment Approaches. Pain Res. Treat. 2011, 2011, 864605. [Google Scholar] [CrossRef]
  25. Valentine, W.M. Toxic Peripheral Neuropathies: Agents and Mechanisms. Toxicol. Pathol. 2020, 48, 152–173. [Google Scholar] [CrossRef]
  26. Cook, A.D.; Christensen, A.D.; Tewari, D.; McMahon, S.B.; Hamilton, J.A. Immune Cytokines and Their Receptors in Inflammatory Pain. Trends Immunol. 2018, 39, 240–255. [Google Scholar] [CrossRef]
  27. Zhang, J.-M.; An, J. Cytokines, Inflammation, and Pain. Int. Anesthesiol. Clin. 2007, 45, 27–37. [Google Scholar] [CrossRef]
  28. Gonçalves dos Santos, G.; Delay, L.; Yaksh, T.L.; Corr, M. Neuraxial Cytokines in Pain States. Front. Immunol. 2020, 10, 3061. [Google Scholar] [CrossRef]
  29. Price, T.J.; Ray, P.R. Recent Advances toward Understanding the Mysteries of the Acute to Chronic Pain Transition. Curr. Opin. Physiol. 2019, 11, 42–50. [Google Scholar] [CrossRef]
  30. Richard, A.M.; Srinivasa, N.R.; James, N.C. Neural Mechanisms of Primary Hyperalgesia. In Neurobiology of Nociceptors; Oxford University Press: Oxford, UK, 1996; pp. 370–389. [Google Scholar]
  31. Ji, R.-R.; Chamessian, A.; Zhang, Y.-Q. Pain Regulation by Non-Neuronal Cells and Inflammation. Science (1979) 2016, 354, 572–577. [Google Scholar] [CrossRef]
  32. Latremoliere, A.; Woolf, C.J. Central Sensitization: A Generator of Pain Hypersensitivity by Central Neural Plasticity. J. Pain 2009, 10, 895–926. [Google Scholar] [CrossRef] [PubMed]
  33. Ashmawi, H.A.; Freire, G.M.G. Peripheral and Central Sensitization. Rev. Dor 2016, 17, 31–34. [Google Scholar] [CrossRef]
  34. Costigan, M.; Woolf, C.J. Pain: Molecular Mechanisms. J. Pain 2000, 1, 35–44. [Google Scholar] [CrossRef] [PubMed]
  35. Tasoulis, T.; Isbister, G. A Review and Database of Snake Venom Proteomes. Toxins 2017, 9, 290. [Google Scholar] [CrossRef] [PubMed]
  36. Cid-Uribe, J.I.; Veytia-Bucheli, J.I.; Romero-Gutierrez, T.; Ortiz, E.; Possani, L.D. Scorpion Venomics: A 2019 Overview. Expert Rev. Proteom. 2020, 17, 67–83. [Google Scholar] [CrossRef] [PubMed]
  37. Langenegger, N.; Nentwig, W.; Kuhn-Nentwig, L. Spider Venom: Components, Modes of Action, and Novel Strategies in Transcriptomic and Proteomic Analyses. Toxins 2019, 11, 611. [Google Scholar] [CrossRef] [PubMed]
  38. Moreno, M.; Giralt, E. Three Valuable Peptides from Bee and Wasp Venoms for Therapeutic and Biotechnological Use: Melittin, Apamin and Mastoparan. Toxins 2015, 7, 1126–1150. [Google Scholar] [CrossRef] [PubMed]
  39. Wehbe, R.; Frangieh, J.; Rima, M.; El Obeid, D.; Sabatier, J.-M.; Fajloun, Z. Bee Venom: Overview of Main Compounds and Bioactivities for Therapeutic Interests. Molecules 2019, 24, 2997. [Google Scholar] [CrossRef]
  40. Carpena, M.; Nuñez-Estevez, B.; Soria-Lopez, A.; Simal-Gandara, J. Bee Venom: An Updating Review of Its Bioactive Molecules and Its Health Applications. Nutrients 2020, 12, 3360. [Google Scholar] [CrossRef]
  41. Clarke, B.T. The natural history of amphibian skin secretions, their normal functioning and potential medical applications. Biol. Rev. 2007, 72, 365–379. [Google Scholar] [CrossRef]
  42. Zhao, Y.; Antunes, A. Biomedical Potential of the Neglected Molluscivorous and Vermivorous Conus Species. Mar. Drugs 2022, 20, 105. [Google Scholar] [CrossRef]
  43. Bjørn-Yoshimoto, W.E.; Ramiro, I.B.L.; Yandell, M.; McIntosh, J.M.; Olivera, B.M.; Ellgaard, L.; Safavi-Hemami, H. Curses or Cures: A Review of the Numerous Benefits Versus the Biosecurity Concerns of Conotoxin Research. Biomedicines 2020, 8, 235. [Google Scholar] [CrossRef]
  44. Tosti, E.; Boni, R.; Gallo, A. Pathophysiological Responses to Conotoxin Modulation of Voltage-Gated Ion Currents. Mar. Drugs 2022, 20, 282. [Google Scholar] [CrossRef]
  45. Hatakeyama, D.M.; Tasima, L.J.; Bravo-Tobar, C.A.; Serino-Silva, C.; Tashima, A.K.; Rodrigues, C.F.; Aguiar, W.D.; Galizio, N.D.; Lima, E.O.; Kavazoi, V.K.; et al. Venom Complexity of Bothrops Atrox (Common Lancehead) Siblings. J. Venom. Anim. Toxins Incl. Trop. Dis. 2020, 26, e20200018. [Google Scholar] [CrossRef]
  46. Valente, R.H.; Guimarães, P.R.; Junqueira, M.; Neves-Ferreira, A.G.C.; Soares, M.R.; Chapeaurouge, A.; Trugilho, M.R.O.; León, I.R.; Rocha, S.L.G.; Oliveira-Carvalho, A.L.; et al. Bothrops Insularis Venomics: A Proteomic Analysis Supported by Transcriptomic-Generated Sequence Data. J. Proteom. 2009, 72, 241–255. [Google Scholar] [CrossRef] [PubMed]
  47. Mora-Obando, D.; Guerrero-Vargas, J.A.; Prieto-Sánchez, R.; Beltrán, J.; Rucavado, A.; Sasa, M.; Gutiérrez, J.M.; Ayerbe, S.; Lomonte, B. Proteomic and Functional Profiling of the Venom of Bothrops Ayerbei from Cauca, Colombia, Reveals Striking Interspecific Variation with Bothrops Asper Venom. J. Proteom. 2014, 96, 159–172. [Google Scholar] [CrossRef] [PubMed]
  48. Sousa, L.F.; Portes-Junior, J.A.; Nicolau, C.A.; Bernardoni, J.L.; Nishiyama, M.Y., Jr.; Amazonas, D.R.; Freitas-de-Sousa, L.A.; Mourão, R.H.; Chalkidis, H.M.; Valente, R.H.; et al. Functional Proteomic Analyses of Bothrops Atrox Venom Reveals Phenotypes Associated with Habitat Variation in the Amazon. J. Proteom. 2017, 159, 32–46. [Google Scholar] [CrossRef] [PubMed]
  49. Simizo, A.; Kitano, E.S.; Sant’Anna, S.S.; Grego, K.F.; Tanaka-Azevedo, A.M.; Tashima, A.K. Comparative Gender Peptidomics of Bothrops Atrox Venoms: Are There Differences between Them? J. Venom. Anim. Toxins Incl. Trop. Dis. 2020, 26, e20200055. [Google Scholar] [CrossRef] [PubMed]
  50. Cavalcante, J.S.; Brito, I.M.; De Oliveira, L.A.; De Barros, L.C.; Almeida, C.; Rossini, B.C.; Sousa, D.L.; Alves, R.S.; Jorge, R.J.; Santos, L.D. Experimental Bothrops Atrox Envenomation: Blood Plasma Proteome Effects after Local Tissue Damage and Perspectives on Thromboinflammation. Toxins 2022, 14, 613. [Google Scholar] [CrossRef]
  51. Cavalcante, J.S.; Borges da Silva, W.R.G.; de Oliveira, L.A.; Brito, I.M.C.; Muller, K.S.; Vidal, I.S.J.; dos Santos, L.D.; Jorge, R.J.B.; Almeida, C.; de Lima Bicho, C. Blood Plasma Proteome Alteration after Local Tissue Damage Induced by Bothrops Erythromelas Snake Venom in Mice. J. Proteom. 2022, 269, 104742. [Google Scholar] [CrossRef] [PubMed]
  52. dos Santos Cavalcante, J.; de Almeida, C.A.; Clasen, M.A.; da Silva, E.L.; de Barros, L.C.; Marinho, A.D.; Rossini, B.C.; Marino, C.L.; Carvalho, P.C.; Jorge, R.J.; et al. A Fingerprint of Plasma Proteome Alteration after Local Tissue Damage Induced by Bothrops Leucurus Snake Venom in Mice. J. Proteom. 2022, 253, 104464. [Google Scholar] [CrossRef] [PubMed]
  53. Sunitha, K.; Hemshekhar, M.; Thushara, R.M.; Santhosh, M.S.; Sundaram, M.S.; Kemparaju, K.; Girish, K.S. Inflammation and Oxidative Stress in Viper Bite: An Insight within and Beyond. Toxicon 2015, 98, 89–97. [Google Scholar] [CrossRef]
  54. Cavalcante, J.D.; de Almeida, D.E.; Moraes, M.S.; Santos, S.R.; Pincinato, P.M.; Riciopo, P.M.; de Oliveira, L.L.; Monteiro, W.M.; Ferreira-Junior, R.S. Challenges and Opportunities in Clinical Diagnostic Routine of Envenomation Using Blood Plasma Proteomics. Toxins 2023, 15, 180. [Google Scholar] [CrossRef]
  55. Cavalcante, J.S.; de Almeida, D.E.G.; Santos-Filho, N.A.; Sartim, M.A.; de Almeida Baldo, A.; Brasileiro, L.; Albuquerque, P.L.; Oliveira, S.S.; Sachett, J.A.G.; Monteiro, W.M.; et al. Crosstalk of Inflammation and Coagulation in Bothrops Snakebite Envenoming: Endogenous Signaling Pathways and Pathophysiology. Int. J. Mol. Sci. 2023, 24, 11508. [Google Scholar] [CrossRef]
  56. Albuquerque, P.L.; Paiva, J.H.; Martins, A.M.; Meneses, G.C.; Silva Júnior, G.B.; Buckley, N.; Daher, E.D. Clinical Assessment and Pathophysiology of Bothrops Venom-Related Acute Kidney Injury: A Scoping Review. J. Venom. Anim. Toxins Incl. Trop. Dis. 2020, 26. [Google Scholar] [CrossRef]
  57. Gutiérrez, J.M.; Calvete, J.J.; Habib, A.G.; Harrison, R.A.; Williams, D.J.; Warrell, D.A. Snakebite Envenoming. Nat. Rev. Dis. Primers 2017, 3, 17063. [Google Scholar] [CrossRef]
  58. Bordon, K.C.F.; Wiezel, G.A.; Amorim, F.G.; Arantes, E.C. Arthropod Venom Hyaluronidases: Biochemical Properties and Potential Applications in Medicine and Biotechnology. J. Venom. Anim. Toxins Incl. Trop. Dis. 2015, 21, 43. [Google Scholar] [CrossRef]
  59. Nait Mohamed, F.A.; Laraba-Djebari, F. Scorpion Envenomation: A Deadly Illness Requiring an Effective Therapy. Toxin Rev. 2021, 40, 592–605. [Google Scholar] [CrossRef]
  60. Schmidt, J.O. Clinical Consequences of Toxic Envenomations by Hymenoptera. Toxicon 2018, 150, 96–104. [Google Scholar] [CrossRef]
  61. RA Mans, D.; Pawirodihardjo, J.; Djotaroeno, M.; Friperson, P. Exploring the Global Animal Biodiversity in the Search for New Drugs -Amphibians. J. Transl. Sci. 2021, 7, 2–17. [Google Scholar] [CrossRef]
  62. Bordon, K.D.; Cologna, C.T.; Fornari-Baldo, E.C.; Pinheiro-Júnior, E.L.; Cerni, F.A.; Amorim, F.G.; Anjolette, F.A.; Cordeiro, F.A.; Wiezel, G.A.; Cardoso, I.A.; et al. From Animal Poisons and Venoms to Medicines: Achievements, Challenges and Perspectives in Drug Discovery. Front. Pharmacol. 2020, 11, 1132. [Google Scholar] [CrossRef]
  63. Trim, S.A.; Trim, C.M. Venom: The Sharp End of Pain Therapeutics. Br. J. Pain 2013, 7, 179–188. [Google Scholar] [CrossRef]
  64. Hayashi, M.A.F.; Oliveira, E.B.; Kerkis, I.; Karpel, R.L. Crotamine: A Novel Cell-Penetrating Polypeptide Nanocarrier with Potential Anti-Cancer and Biotechnological Applications. Methods Mol. Biol. 2012, 906, 337–352. [Google Scholar] [CrossRef]
  65. Stábeli, R.G.; Marcussi, S.; Carlos, G.B.; Pietro, R.C.L.R.; Selistre-De-Araújo, H.S.; Giglio, J.R.; Oliveira, E.B.; Soares, A.M. Platelet Aggregation and Antibacterial Effects of an L-Amino Acid Oxidase Purified from Bothrops Alternatus Snake Venom. Bioorg Med. Chem. 2004, 12, 2881–2886. [Google Scholar] [CrossRef]
  66. Abbade, L.P.; Barraviera, S.R.; Silvares, M.R.; Lima, A.B.; Haddad, G.R.; Gatti, M.A.; Medolago, N.B.; Rigotto Carneiro, M.T.; Dos Santos, L.D.; Ferreira, R.S., Jr.; et al. Treatment of Chronic Venous Ulcers With Heterologous Fibrin Sealant: A Phase I/II Clinical Trial. Front. Immunol. 2021, 12, 83. [Google Scholar] [CrossRef]
  67. FERREIRA, S.H. A Bradykinin-Potentiating Factor (Bpf) Present in the Venom of Bothrops Jararaca. Br. J. Pharmacol. Chemother. 1965, 24, 163. [Google Scholar] [CrossRef]
  68. Diochot, S.; Alloui, A.; Rodrigues, P.; Dauvois, M.; Friend, V.; Aissouni, Y.; Eschalier, A.; Lingueglia, E.; Baron, A. Analgesic Effects of Mambalgin Peptide Inhibitors of Acid-Sensing Ion Channels in Inflammatory and Neuropathic Pain. Pain 2016, 157, 552–559. [Google Scholar] [CrossRef]
  69. Zhang, F.; Zhang, C.; Xu, X.; Zhang, Y.; Gong, X.; Yang, Z.; Zhang, H.; Tang, D.; Liang, S.; Liu, Z. Naja Atra Venom Peptide Reduces Pain by Selectively Blocking the Voltage-Gated Sodium Channel Nav1.8. J. Biol. Chem. 2019, 294, 7324–7334. [Google Scholar] [CrossRef]
  70. Sant’Anna, M.B.; Lopes, F.S.R.; Kimura, L.F.; Giardini, A.C.; Sant’Anna, O.A.; Picolo, G. Crotoxin Conjugated to SBA-15 Nanostructured Mesoporous Silica Induces Long-Last Analgesic Effect in the Neuropathic Pain Model in Mice. Toxins 2019, 11, 679. [Google Scholar] [CrossRef]
  71. Teixeira, N.B.; Sant’Anna, M.B.; Giardini, A.C.; Araujo, L.P.; Fonseca, L.A.; Basso, A.S.; Cury, Y.; Picolo, G. Crotoxin Down-Modulates pro-Inflammatory Cells and Alleviates Pain on the MOG35-55-Induced Experimental Autoimmune Encephalomyelitis, an Animal Model of Multiple Sclerosis. Brain Behav. Immun. 2020, 84, 253–268. [Google Scholar] [CrossRef]
  72. Konno, K.; Picolo, G.; Gutierrez, V.P.; Brigatte, P.; Zambelli, V.O.; Camargo, A.C.M.; Cury, Y. Crotalphine, a Novel Potent Analgesic Peptide from the Venom of the South American Rattlesnake Crotalus Durissus Terrificus. Peptides 2008, 29, 1293–1304. [Google Scholar] [CrossRef]
  73. Bressan, E.; Touska, F.; Vetter, I.; Kistner, K.; Kichko, T.I.; Teixeira, N.B.; Picolo, G.; Cury, Y.; Lewis, R.J.; Fischer, M.J.M.; et al. Crotalphine Desensitizes TRPA1 Ion Channels to Alleviate Inflammatory Hyperalgesia. Pain 2016, 157, 2504–2516. [Google Scholar] [CrossRef] [PubMed]
  74. Zhao, C.; Zhao, J.; Yang, Q.; Ye, Y. Cobra Neurotoxin Produces Central Analgesic and Hyperalgesic Actions via Adenosine A 1 and A 2A Receptors. Mol. Pain 2017, 13, 174480691772033. [Google Scholar] [CrossRef]
  75. Liang, Y.-X.; Zhang, Z.-Y.; Zhang, R. Antinociceptive Effect of Najanalgesin from Naja Naja Atra in a Neuropathic Pain Model via Inhibition of C-Jun NH2-Terminal Kinase. Chin. Med. J. 2015, 128, 2340–2345. [Google Scholar] [CrossRef]
  76. Gong, S.; Liang, Q.; Zhu, Q.; Ding, D.; Yin, Q.; Tao, J.; Jiang, X. Nicotinic Acetylcholine Receptor A7 Subunit Is Involved in the Cobratoxin-Induced Antinociception in an Animal Model of Neuropathic Pain. Toxicon 2015, 93, 31–36. [Google Scholar] [CrossRef]
  77. Kampo, S.; Ahmmed, B.; Zhou, T.; Owusu, L.; Anabah, T.W.; Doudou, N.R.; Kuugbee, E.D.; Cui, Y.; Lu, Z.; Yan, Q.; et al. Scorpion Venom Analgesic Peptide, BmK AGAP Inhibits Stemness, and Epithelial-Mesenchymal Transition by Down-Regulating PTX3 in Breast Cancer. Front. Oncol. 2019, 9, 21. [Google Scholar] [CrossRef]
  78. Cao, Q.; Lu, W.; Cai, X.; Hu, C.; Wang, C.; Ye, J.; Yan, H.; Yang, Y.; Wang, Z.; Huo, J.; et al. In Vitro Refolding and Functional Analysis of Polyhistidine-Tagged Buthus Martensii Karsch Antitumor-Analgesic Peptide Produced in Escherichia Coli. Biotechnol. Lett. 2015, 37, 2461–2466. [Google Scholar] [CrossRef]
  79. Xu, Y.; Meng, X.; Hou, X.; Sun, J.; Kong, X.; Sun, Y.; Liu, Z.; Ma, Y.; Niu, Y.; Song, Y.; et al. A Mutant of the Buthus Martensii Karsch Antitumor-Analgesic Peptide Exhibits Reduced Inhibition to HNav1.4 and HNav1.5 Channels While Retaining Analgesic Activity. J. Biol. Chem. 2017, 292, 18270–18280. [Google Scholar] [CrossRef]
  80. Ruan, J.-P.; Mao, Q.-H.; Lu, W.-G.; Cai, X.-T.; Chen, J.; Li, Q.-; Fu, Q.-; Yan, H.-J.; Cao, J.-L.; Cao, P. Inhibition of Spinal MAPKs by Scorpion Venom Peptide BmK AGAP Produces a Sensory-Specific Analgesic Effect. Mol. Pain 2018, 14, 174480691876123. [Google Scholar] [CrossRef]
  81. Maatoug, R.; Jebali, J.; Guieu, R.; De Waard, M.; Kharrat, R. BotAF, a New Buthus Occitanus Tunetanus Scorpion Toxin, Produces Potent Analgesia in Rodents. Toxicon 2018, 149, 72–85. [Google Scholar] [CrossRef]
  82. Li, Z.; Hu, P.; Wu, W.; Wang, Y. Peptides with Therapeutic Potential in the Venom of the Scorpion Buthus Martensii Karsch. Peptides 2019, 115, 43–50. [Google Scholar] [CrossRef] [PubMed]
  83. Rigo, F.K.; Bochi, G.V.; Pereira, A.L.; Adamante, G.; Ferro, P.R.; Dal-Toé De Prá, S.; Milioli, A.M.; Damiani, A.P.; da Silveira Prestes, G.; Dalenogare, D.P.; et al. TsNTxP, a Non-Toxic Protein from Tityus Serrulatus Scorpion Venom, Induces Antinociceptive Effects by Suppressing Glutamate Release in Mice. Eur. J. Pharmacol. 2019, 855, 65–74. [Google Scholar] [CrossRef]
  84. Bagheri-Ziari, S.; Shahbazzadeh, D.; Sardari, S.; Sabatier, J.-M.; Pooshang Bagheri, K. Discovery of a New Analgesic Peptide, Leptucin, from the Iranian Scorpion, Hemiscorpius Lepturus. Molecules 2021, 26, 2580. [Google Scholar] [CrossRef] [PubMed]
  85. Cui, Y.; Wang, Y.; Wang, X.; Zhang, Z.; Zhang, J.; Zhao, Y. The Role of the Arginine Residue in Site RC for the Analgesic Activity of the Recombinant Chinese Scorpion Buthus Martensii Karsch, BmK AGP-SYPU1. Comput. Biol. Chem. 2018, 74, 247–252. [Google Scholar] [CrossRef]
  86. Chen, Y.; Xu, E.; Sang, M.; Wang, Z.; Zhang, Y.; Ye, J.; Zhou, Q.; Zhao, C.; Hu, C.; Lu, W.; et al. Makatoxin-3, a Thermostable Nav1.7 Agonist from Buthus Martensii Karsch (BmK) Scorpion Elicits Non-Narcotic Analgesia in Inflammatory Pain Models. J. Ethnopharmacol. 2022, 288, 114998. [Google Scholar] [CrossRef] [PubMed]
  87. Sousa, S.R.; Wingerd, J.S.; Brust, A.; Bladen, C.; Ragnarsson, L.; Herzig, V.; Deuis, J.R.; Dutertre, S.; Vetter, I.; Zamponi, G.W.; et al. Discovery and Mode of Action of a Novel Analgesic β-Toxin from the African Spider Ceratogyrus Darlingi. PLoS ONE 2017, 12, e0182848. [Google Scholar] [CrossRef] [PubMed]
  88. Cardoso, F.C.; Dekan, Z.; Smith, J.J.; Deuis, J.R.; Vetter, I.; Herzig, V.; Alewood, P.F.; King, G.F.; Lewis, R.J. Modulatory Features of the Novel Spider Toxin Μ-TRTX-Df1a Isolated from the Venom of the Spider Davus Fasciatus. Br. J. Pharmacol. 2017, 174, 2528–2544. [Google Scholar] [CrossRef] [PubMed]
  89. Meng, P.; Huang, H.; Wang, G.; Yang, S.; Lu, Q.; Liu, J.; Lai, R.; Rong, M. A Novel Toxin from Haplopelma Lividum Selectively Inhibits the NaV1.8 Channel and Possesses Potent Analgesic Efficacy. Toxins 2016, 9, 7. [Google Scholar] [CrossRef]
  90. Wu, X.; Wang, Z.; Chen, Y.; Xu, D.; Zhang, P.; Wang, X. Newly Discovered Action of HpTx3 from Venom of Heteropoda Venatoria on Nav1.7 and Its Pharmacological Implications in Analgesia. Toxins 2019, 11, 680. [Google Scholar] [CrossRef]
  91. Cardoso, F.C.; Dekan, Z.; Rosengren, K.J.; Erickson, A.; Vetter, I.; Deuis, J.R.; Herzig, V.; Alewood, P.F.; King, G.F.; Lewis, R.J. Identification and Characterization of ProTx-III [μ -TRTX-Tp1a], a New Voltage-Gated Sodium Channel Inhibitor from Venom of the Tarantula Thrixopelma Pruriens. Mol. Pharmacol. 2015, 88, 291–303. [Google Scholar] [CrossRef]
  92. da Silva, J.F.; Castro-Junior, C.J.; Oliveira, S.M.; Dalmolin, G.D.; Silva, C.R.; Vieira, L.B.; Diniz, D.M.; do Nascimento Cordeiro, M.; Ferreira, J.; Souza, A.H.; et al. Characterization of the Antinociceptive Effect of PhTx3-4, a Toxin from Phoneutria Nigriventer, in Models of Thermal, Chemical and Incisional Pain in Mice. Toxicon 2015, 108, 53–61. [Google Scholar] [CrossRef]
  93. Emerich, B.; Ferreira, R.; Cordeiro, M.; Borges, M.; Pimenta, A.; Figueiredo, S.; Duarte, I.; de Lima, M. δ-Ctenitoxin-Pn1a, a Peptide from Phoneutria Nigriventer Spider Venom, Shows Antinociceptive Effect Involving Opioid and Cannabinoid Systems, in Rats. Toxins 2016, 8, 106. [Google Scholar] [CrossRef]
  94. Freitas, A.C.N.; Pacheco, D.F.; Machado, M.F.M.; Carmona, A.K.; Duarte, I.D.G.; de Lima, M.E. PnPP-19, a Spider Toxin Peptide, Induces Peripheral Antinociception through Opioid and Cannabinoid Receptors and Inhibition of Neutral Endopeptidase. Br. J. Pharmacol. 2016, 173, 1491–1501. [Google Scholar] [CrossRef]
  95. Freitas, A.C.N.; Silva, G.C.; Pacheco, D.F.; Pimenta, A.M.C.; Lemos, V.S.; Duarte, I.D.G.; de Lima, M.E. The Synthetic Peptide PnPP-19 Induces Peripheral Antinociception via Activation of NO/CGMP/KATP Pathway: Role of ENOS and NNOS. Nitric Oxide 2017, 64, 31–38. [Google Scholar] [CrossRef]
  96. Stanciu, G.-D.; Luca, A.; Marza, A.; Alexa-Stratulat, T.; Tudorancea, I.; Bild, W.; Rezus, E.; Rezus, C.; Tamba, B.I. Intracerebroventricular Coadministration of Protoxin-II and Trace Elements in Rats Enhances the Analgesic Effect of the 1.7 Voltage-Gate Sodium Channel Blocker. Biomed. Res. Int. 2019, 2019, 8057803. [Google Scholar] [CrossRef]
  97. Rigo, F.K.; Trevisan, G.; De Prá, S.D.-T.; Cordeiro, M.N.; Borges, M.H.; Silva, J.F.; Santa Cecilia, F.V.; de Souza, A.H.; de Oliveira Adamante, G.; Milioli, A.M.; et al. The Spider Toxin Phα1β Recombinant Possesses Strong Analgesic Activity. Toxicon 2017, 133, 145–152. [Google Scholar] [CrossRef]
  98. da Silva Junior, C.A.; de Castro Junior, C.J.; Pereira, E.M.R.; Binda, N.S.; da Silva, J.F.; do Nascimento Cordeiro, M.; Diniz, D.M.; Cecilia, F.S.; Ferreira, J.; Gomez, M.V. The Inhibitory Effect of Phα1β Toxin on Diabetic Neuropathic Pain Involves the CXCR4 Chemokine Receptor. Pharmacol. Rep. 2020, 72, 47–54. [Google Scholar] [CrossRef]
  99. Deuis, J.R.; Dekan, Z.; Wingerd, J.S.; Smith, J.J.; Munasinghe, N.R.; Bhola, R.F.; Imlach, W.L.; Herzig, V.; Armstrong, D.A.; Rosengren, K.J.; et al. Pharmacological Characterisation of the Highly NaV1.7 Selective Spider Venom Peptide Pn3a. Sci. Rep. 2017, 7, 40883. [Google Scholar] [CrossRef]
  100. Deng, M.; Jiang, L.; Luo, X.; Tao, H.; Liang, S. Jingzhaotoxin-X, a Gating Modifier of Kv4.2 and Kv4.3 Potassium Channels Purified from the Venom of the Chinese Tarantula Chilobrachys Jingzhao. J. Venom. Anim. Toxins Incl. Trop. Dis. 2020, 26, e20190043. [Google Scholar] [CrossRef] [PubMed]
  101. Zhang, Y.; Peng, D.; Huang, B.; Yang, Q.; Zhang, Q.; Chen, M.; Rong, M.; Liu, Z. Discovery of a Novel Nav1.7 Inhibitor From Cyriopagopus Albostriatus Venom With Potent Analgesic Efficacy. Front. Pharmacol. 2018, 9, 1158. [Google Scholar] [CrossRef] [PubMed]
  102. Zhang, Y.; Peng, D.; Zhang, Q.; Huang, B.; Yang, Q.; Tang, D.; Chen, M.; Rong, M.; Liu, Z. Μ-TRTX-Ca1a: A Novel Neurotoxin from Cyriopagopus Albostriatus with Analgesic Effects. Acta Pharmacol. Sin. 2019, 40, 859–866. [Google Scholar] [CrossRef]
  103. Gonçalves, T.C.; Benoit, E.; Kurz, M.; Lucarain, L.; Fouconnier, S.; Combemale, S.; Jaquillard, L.; Schombert, B.; Chambard, J.; Boukaiba, R.; et al. From Identification to Functional Characterization of Cyriotoxin-1a, an Antinociceptive Toxin from the Spider Cyriopagopus schioedtei. Br. J. Pharmacol. 2019, 176, 1298–1314. [Google Scholar] [CrossRef]
  104. Chen, C.; Xu, B.; Shi, X.; Zhang, M.; Zhang, Q.; Zhang, T.; Zhao, W.; Zhang, R.; Wang, Z.; Li, N.; et al. GpTx-1 and [Ala5, Phe6, Leu26, Arg28]GpTx-1, Two Peptide Na V 1.7 Inhibitors: Analgesic and Tolerance Properties at the Spinal Level. Br. J. Pharmacol. 2018, 175, 3911–3927. [Google Scholar] [CrossRef]
  105. Oliveira, S.M.; Silva, C.R.; Trevisan, G.; Villarinho, J.G.; Cordeiro, M.N.; Richardson, M.; Borges, M.H.; Castro, C.J.; Gomez, M.V.; Ferreira, J. Antinociceptive Effect of a Novel Armed Spider Peptide Tx3-5 in Pathological Pain Models in Mice. Pflug. Arch. 2016, 468, 881–894. [Google Scholar] [CrossRef]
  106. Tonello, R.; Fusi, C.; Materazzi, S.; Marone, I.M.; De Logu, F.; Benemei, S.; Gonçalves, M.C.; Coppi, E.; Castro-Junior, C.J.; Gomez, M.V.; et al. The Peptide Phα1β, from Spider Venom, Acts as a TRPA1 Channel Antagonist with Antinociceptive Effects in Mice. Br. J. Pharmacol. 2017, 174, 57–69. [Google Scholar] [CrossRef]
  107. Oliveira, C.F.; Alves, D.P.; Emerich, B.L.; Figueiredo, S.G.; Cordeiro, M.D.; Borges, M.H.; Richardson, M.; Pimenta, A.M.; Duarte, I.D.; Lima, M.E. Antinociceptive Effect of PnTx4(5-5), a Peptide from Phoneutria Nigriventer Spider Venom, in Rat Models and the Involvement of Glutamatergic System. J. Venom. Anim. Toxins Incl. Trop. Dis. 2019, 25, e20190022. [Google Scholar] [CrossRef]
  108. Rigo, F.K.; Rossato, M.F.; Borges, V.; Silva, J.F.; Pereira, E.M.; Ávila, R.A.; Trevisan, G.; Santos, D.C.; Diniz, D.M.; Silva, M.A.; et al. Analgesic and Side Effects of Intravenous Recombinant Phα1β. J. Venom. Anim. Toxins Incl. Trop. Dis. 2020, 26, e20190070. [Google Scholar] [CrossRef]
  109. Antunes, F.T.T.; Angelo, S.G.; Dallegrave, E.; Picada, J.N.; Marroni, N.P.; Schemitt, E.; Ferraz, A.G.; Gomez, M.V.; de Souza, A.H. Recombinant Peptide Derived from the Venom the Phoneutria Nigriventer Spider Relieves Nociception by Nerve Deafferentation. Neuropeptides 2020, 79, 101980. [Google Scholar] [CrossRef]
  110. Emerich, B.L.; Ferreira, R.C.M.; Machado-de-Avila, R.A.; Resende, J.M.; Duarte, I.D.G.; de Lima, M.E. PnAn13, an Antinociceptive Synthetic Peptide Inspired in the Phoneutria Nigriventer Toxin PnTx4(6–1) (δ-Ctenitoxin-Pn1a). Toxicon X 2020, 7, 100045. [Google Scholar] [CrossRef]
  111. Tonello, R.; Trevisan, G.; Luckemeyer, D.; Castro-Junior, C.J.; Gomez, M.V.; Ferreira, J. Phα1β, a Dual Blocker of TRPA1 and Cav2.2, as an Adjuvant Drug in Opioid Therapy for Postoperative Pain. Toxicon 2020, 188, 80–88. [Google Scholar] [CrossRef] [PubMed]
  112. Garcia Mendes, M.P.; Carvalho dos Santos, D.; Rezende, M.J.S.; Assis Ferreira, L.C.; Rigo, F.K.; José de Castro Junior, C.; Gomez, M.V. Effects of Intravenous Administration of Recombinant Phα1β Toxin in a Mouse Model of Fibromyalgia. Toxicon 2021, 195, 104–110. [Google Scholar] [CrossRef]
  113. Dornelles, M.F.; Marques, M.G.B.; Renner, M.F. Revisão Sobre Toxinas de Anura (Tetrapoda, Lissamphibia) e Suas Aplicações Biotecnológicas. Ciência Em Mov. 2010, 12, 103–114. [Google Scholar] [CrossRef]
  114. Tao, J.; Jiang, F.; Liu, C.; Liu, Z.; Zhu, Y.; Xu, J.; Ge, Y.; Xu, K.; Yin, P. Modulatory Effects of Bufalin, an Active Ingredient from Toad Venom on Voltage-Gated Sodium Channels. Mol. Biol. Rep. 2018, 45, 721–740. [Google Scholar] [CrossRef]
  115. Pucca, M.B.; Cerni, F.A.; Oliveira, I.S.; Jenkins, T.P.; Argemí, L.; Sørensen, C.V.; Ahmadi, S.; Barbosa, J.E.; Laustsen, A.H. Bee Updated: Current Knowledge on Bee Venom and Bee Envenoming Therapy. Front. Immunol. 2019, 10, 2090. [Google Scholar] [CrossRef]
  116. Kang, S.-Y.; Roh, D.-H.; Choi, J.-W.; Ryu, Y.; Lee, J.-H. Repetitive Treatment with Diluted Bee Venom Attenuates the Induction of Below-Level Neuropathic Pain Behaviors in a Rat Spinal Cord Injury Model. Toxins 2015, 7, 2571–2585. [Google Scholar] [CrossRef]
  117. Yeo, J.-H.; Yoon, S.-Y.; Kwon, S.-K.; Kim, S.-J.; Lee, J.-H.; Beitz, A.J.; Roh, D.-H. Repetitive Acupuncture Point Treatment with Diluted Bee Venom Relieves Mechanical Allodynia and Restores Intraepidermal Nerve Fiber Loss in Oxaliplatin-Induced Neuropathic Mice. J. Pain 2016, 17, 298–309. [Google Scholar] [CrossRef]
  118. Lee, S.; Lee, J.; Kim, Y.; Choi, J.; Jeon, S.; Kim, D.; Jeong, H.; Lee, Y.; Park, H. Antiallodynic Effects of Bee Venom in an Animal Model of Complex Regional Pain Syndrome Type 1 (CRPS-I). Toxins 2017, 9, 285. [Google Scholar] [CrossRef]
  119. Lee, J.H.; Gang, J.; Yang, E.; Kim, W.; Jin, Y.-H. Bee Venom Acupuncture Attenuates Oxaliplatin-Induced Neuropathic Pain by Modulating Action Potential Threshold in A-Fiber Dorsal Root Ganglia Neurons. Toxins 2020, 12, 737. [Google Scholar] [CrossRef]
  120. Li, D.; Chung, G.; Kim, S.K. The Involvement of Central Noradrenergic Pathway in the Analgesic Effect of Bee Venom Acupuncture on Vincristine-Induced Peripheral Neuropathy in Rats. Toxins 2020, 12, 775. [Google Scholar] [CrossRef]
  121. Kang, D.-W.; Choi, J.-G.; Kim, J.; Park, J.B.; Lee, J.-H.; Kim, H.-W. Bee Venom Reduces Burn-Induced Pain via the Suppression of Peripheral and Central Substance P Expression in Mice. J. Vet. Sci. 2021, 22, e9. [Google Scholar] [CrossRef]
  122. Choi, J.; Jeon, C.; Lee, J.; Jang, J.; Quan, F.; Lee, K.; Kim, W.; Kim, S. Suppressive Effects of Bee Venom Acupuncture on Paclitaxel-Induced Neuropathic Pain in Rats: Mediation by Spinal A2-Adrenergic Receptor. Toxins 2017, 9, 351. [Google Scholar] [CrossRef]
  123. Li, D.; Lee, Y.; Kim, W.; Lee, K.; Bae, H.; Kim, S. Analgesic Effects of Bee Venom Derived Phospholipase A2 in a Mouse Model of Oxaliplatin-Induced Neuropathic Pain. Toxins 2015, 7, 2422–2434. [Google Scholar] [CrossRef]
  124. El-tedawy, D.; Abd-alhaseeb, M.; Helmy, M.; Ghoneim, A. Systemic Bee Venom Exerts Anti-arthritic and Anti-inflammatory Properties in a Rat Model of Arthritis. Biomed. Rep. 2020, 13, 20. [Google Scholar] [CrossRef]
  125. Kim, W.; Kim, M.; Go, D.; Min, B.-I.; Na, H.; Kim, S. Combined Effects of Bee Venom Acupuncture and Morphine on Oxaliplatin-Induced Neuropathic Pain in Mice. Toxins 2016, 8, 33. [Google Scholar] [CrossRef]
  126. Li, D.; Yoo, J.H.; Kim, S.K. Long-Lasting and Additive Analgesic Effects of Combined Treatment of Bee Venom Acupuncture and Venlafaxine on Paclitaxel-Induced Allodynia in Mice. Toxins 2020, 12, 620. [Google Scholar] [CrossRef] [PubMed]
  127. Lin, L.; Zhu, B.-P.; Cai, L. Therapeutic Effect of Melittin on a Rat Model of Chronic Prostatitis Induced by Complete Freund’s Adjuvant. Biomed. Pharmacother. 2017, 90, 921–927. [Google Scholar] [CrossRef] [PubMed]
  128. Li, D.; Kim, W.; Shin, D.; Jung, Y.; Bae, H.; Kim, S. Preventive Effects of Bee Venom Derived Phospholipase A2 on Oxaliplatin-Induced Neuropathic Pain in Mice. Toxins 2016, 8, 27. [Google Scholar] [CrossRef] [PubMed]
  129. Safavi-Hemami, H.; Brogan, S.E.; Olivera, B.M. Pain Therapeutics from Cone Snail Venoms: From Ziconotide to Novel Non-Opioid Pathways. J. Proteom. 2019, 190, 12–20. [Google Scholar] [CrossRef]
  130. Gorson, J.; Holford, M. Small Packages, Big Returns: Uncovering the Venom Diversity of Small Invertebrate Conoidean Snails. Integr. Comp. Biol. 2016, 56, 962–972. [Google Scholar] [CrossRef]
  131. Yu, S.; Du, T.; Liu, Z.; Wu, Q.; Feng, G.; Dong, M.; Zhou, X.; Jiang, L.; Dai, Q. Im10A, a Short Conopeptide Isolated from Conus Imperialis and Possesses Two Highly Concentrated Disulfide Bridges and Analgesic Activity. Peptides 2016, 81, 15–20. [Google Scholar] [CrossRef] [PubMed]
  132. Yang, M.; Zhao, S.; Min, X.; Shao, M.; Chen, Y.; Chen, Z.; Zhou, M. A Novel μ-Conotoxin from Worm-Hunting Conus Tessulatus That Selectively Inhibit Rat TTX-Resistant Sodium Currents. Toxicon 2017, 130, 11–18. [Google Scholar] [CrossRef]
  133. Liu, Z.; Bartels, P.; Sadeghi, M.; Du, T.; Dai, Q.; Zhu, C.; Yu, S.; Wang, S.; Dong, M.; Sun, T.; et al. A Novel α-Conopeptide Eu1.6 Inhibits N-Type (CaV2.2) Calcium Channels and Exhibits Potent Analgesic Activity. Sci. Rep. 2018, 8, 1004. [Google Scholar] [CrossRef]
  134. Sousa, S.R.; McArthur, J.R.; Brust, A.; Bhola, R.F.; Rosengren, K.J.; Ragnarsson, L.; Dutertre, S.; Alewood, P.F.; Christie, M.J.; Adams, D.J.; et al. Novel Analgesic ω-Conotoxins from the Vermivorous Cone Snail Conus Moncuri Provide New Insights into the Evolution of Conopeptides. Sci. Rep. 2018, 8, 13397. [Google Scholar] [CrossRef] [PubMed]
  135. Cai, F.; Xu, N.; Liu, Z.; Ding, R.; Yu, S.; Dong, M.; Wang, S.; Shen, J.; Tae, H.-S.; Adams, D.J.; et al. Targeting of N-Type Calcium Channels via GABA B -Receptor Activation by α-Conotoxin Vc1.1 Variants Displaying Improved Analgesic Activity. J. Med. Chem. 2018, 61, 10198–10205. [Google Scholar] [CrossRef] [PubMed]
  136. Belgi, A.; Burnley, J.V.; MacRaild, C.A.; Chhabra, S.; Elnahriry, K.A.; Robinson, S.D.; Gooding, S.G.; Tae, H.-S.; Bartels, P.; Sadeghi, M.; et al. Alkyne-Bridged α-Conotoxin Vc1.1 Potently Reverses Mechanical Allodynia in Neuropathic Pain Models. J. Med. Chem. 2021, 64, 3222–3233. [Google Scholar] [CrossRef] [PubMed]
  137. Castro, J.; Grundy, L.; Deiteren, A.; Harrington, A.M.; O’Donnell, T.; Maddern, J.; Moore, J.; Garcia-Caraballo, S.; Rychkov, G.Y.; Yu, R.; et al. Cyclic Analogues of A-conotoxin Vc1.1 Inhibit Colonic Nociceptors and Provide Analgesia in a Mouse Model of Chronic Abdominal Pain. Br. J. Pharmacol. 2018, 175, 2384–2398. [Google Scholar] [CrossRef] [PubMed]
  138. Wang, H.; Li, X.; Zhangsun, D.; Yu, G.; Su, R.; Luo, S. The α9α10 Nicotinic Acetylcholine Receptor Antagonist AO-Conotoxin GeXIVA[1,2] Alleviates and Reverses Chemotherapy-Induced Neuropathic Pain. Mar. Drugs 2019, 17, 265. [Google Scholar] [CrossRef]
  139. Liu, C.; Wu, P.; Zhu, H.; Grieco, P.; Yu, R.; Gao, X.; Wu, G.; Wang, D.; Xu, H.; Qi, W. Rationally Designed α-Conotoxin Analogues Maintained Analgesia Activity and Weakened Side Effects. Molecules 2019, 24, 337. [Google Scholar] [CrossRef]
  140. Gajewiak, J.; Christensen, S.B.; Dowell, C.; Hararah, F.; Fisher, F.; Huynh, P.N.; Olivera, B.M.; McIntosh, J.M. Selective Penicillamine Substitution Enables Development of a Potent Analgesic Peptide That Acts through a Non-Opioid-Based Mechanism. J. Med. Chem. 2021, 64, 9271–9278. [Google Scholar] [CrossRef]
  141. Hasan, M.d.M.; Starobova, H.; Mueller, A.; Vetter, I.; Lewis, R.J. Subcutaneous ω-Conotoxins Alleviate Mechanical Pain in Rodent Models of Acute Peripheral Neuropathy. Mar. Drugs 2021, 19, 106. [Google Scholar] [CrossRef]
  142. Liu, X.; Yao, G.; Wang, K.; Liu, Y.; Wan, X.; Jiang, H. Structural and Functional Characterization of Conotoxins from Conus Achatinus Targeting NMDAR. Mar. Drugs 2020, 18, 135. [Google Scholar] [CrossRef]
  143. Zheng, N.; Christensen, S.B.; Dowell, C.; Purushottam, L.; Skalicky, J.J.; McIntosh, J.M.; Chou, D.H.-C. Discovery of Methylene Thioacetal-Incorporated α-RgIA Analogues as Potent and Stable Antagonists of the Human A9α10 Nicotinic Acetylcholine Receptor for the Treatment of Neuropathic Pain. J. Med. Chem. 2021, 64, 9513–9524. [Google Scholar] [CrossRef] [PubMed]
  144. Yu, S.; Li, Y.; Chen, J.; Zhang, Y.; Tao, X.; Dai, Q.; Wang, Y.; Li, S.; Dong, M. TAT-Modified ω-Conotoxin MVIIA for Crossing the Blood-Brain Barrier. Mar. Drugs 2019, 17, 286. [Google Scholar] [CrossRef] [PubMed]
  145. Rajabi, H.; Zolgharnein, H.; Ronagh, M.T.; Moghaddam, J.A.; Crüsemann, M. Conus Coronatus and Conus Frigidus Venom: A New Source of Conopeptides with Analgesic Activity. Avicenna J. Med. Biotechnol. 2020, 12, 179–185. [Google Scholar] [PubMed]
  146. Chen, J.; Liu, X.; Yu, S.; Liu, J.; Chen, R.; Zhang, Y.; Jiang, L.; Dai, Q. A Novel ω-Conotoxin Bu8 Inhibiting N-Type Voltage-Gated Calcium Channels Displays Potent Analgesic Activity. Acta Pharm. Sin. B 2021, 11, 2685–2693. [Google Scholar] [CrossRef] [PubMed]
  147. Qiang, Y.; Wu, Y.; Zhao, D.; Zhao, B.; Wang, F.; Ren, S.; Wen, Y.; Gu, J.; Zhang, L.; Liu, K.; et al. Discovery and Characterization of the Novel Conotoxin Lv1d from Conus Lividus That Presents Analgesic Activity. Toxicon 2021, 194, 70–78. [Google Scholar] [CrossRef] [PubMed]
  148. Qiang, Y.; Niu, J.; Wu, Y.; Di, Z.; Wang, F.; Zhang, L.; Liu, K.; Zhao, B.; Wang, L. Discovery of a Novel Cysteine Framework XXIV Conotoxin from Conus Striatus, S24a, with Potential Analgesic Activity. Int. J. Pept. Res. Ther. 2021, 27, 615–625. [Google Scholar] [CrossRef]
  149. Mohamed, A.A.; Nabil, Z.I.; El-Naggar, M.S. Prospecting for Candidate Molecules from Conus Virgo Toxins to Develop New Biopharmaceuticals. J. Venom. Anim. Toxins Incl. Trop. Dis. 2022, 28. [Google Scholar] [CrossRef]
  150. Williams, J.A.; Day, M.; Heavner, J.E. Ziconotide: An Update and Review. Expert. Opin. Pharmacother. 2008, 9, 1575–1583. [Google Scholar] [CrossRef]
  151. Ibraheem, D.; Elaissari, A.; Fessi, H. Administration Strategies for Proteins and Peptides. Int. J. Pharm. 2014, 477, 578–589. [Google Scholar] [CrossRef]
  152. Abram, S.E.; Mampilly, G.A.; Milosavljevic, D. Assessment of the Potency and Intrinsic Activity of Systemic versus Intrathecal Opioids in Rats. Anesthesiology 1997, 87, 127–134. [Google Scholar] [CrossRef]
  153. Mercadante, S. Problems of Long-Term Spinal Opioid Treatment in Advanced Cancer Patients. Pain 1999, 79, 1–13. [Google Scholar] [CrossRef]
  154. Morishita, M.; Peppas, N.A. Is the Oral Route Possible for Peptide and Protein Drug Delivery? Drug Discov. Today 2006, 11, 905–910. [Google Scholar] [CrossRef]
  155. Craik, D.J.; Fairlie, D.P.; Liras, S.; Price, D. The Future of Peptide-Based Drugs. Chem. Biol. Drug Des. 2013, 81, 136–147. [Google Scholar] [CrossRef] [PubMed]
  156. Lipinski, C.A. Drug-like Properties and the Causes of Poor Solubility and Poor Permeability. J. Pharmacol. Toxicol. Methods 2000, 44, 235–249. [Google Scholar] [CrossRef] [PubMed]
  157. Muttenthaler, M.; King, G.F.; Adams, D.J.; Alewood, P.F. Trends in Peptide Drug Discovery. Nat. Rev. Drug Discov. 2021, 20, 309–325. [Google Scholar] [CrossRef] [PubMed]
  158. Tan, M.X.L.; Danquah, M.K. Drug and Protein Encapsulation by Emulsification: Technology Enhancement Using Foam Formulations. Chem. Eng. Technol. 2012, 35, 618–626. [Google Scholar] [CrossRef]
  159. Angelova, A.; Angelov, B.; Drechsler, M.; Garamus, V.M.; Lesieur, S. Protein Entrapment in PEGylated Lipid Nanoparticles. Int. J. Pharm. 2013, 454, 625–632. [Google Scholar] [CrossRef] [PubMed]
  160. Mehnert, W. Solid Lipid Nanoparticles Production, Characterization and Applications. Adv. Drug Deliv. Rev. 2001, 47, 165–196. [Google Scholar] [CrossRef]
  161. Swaminathan, J.; Ehrhardt, C. Liposomal Delivery of Proteins and Peptides. Expert. Opin. Drug Deliv. 2012, 9, 1489–1503. [Google Scholar] [CrossRef]
  162. Wang, Y.; Selomulya, C. Spray Drying Strategy for Encapsulation of Bioactive Peptide Powders for Food Applications. Adv. Powder Technol. 2020, 31, 409–415. [Google Scholar] [CrossRef]
  163. Ganesh, A.N.; Heusser, C.; Garad, S.; Sánchez-Félix, M.V. Patient-Centric Design for Peptide Delivery: Trends in Routes of Administration and Advancement in Drug Delivery Technologies. Med. Drug Discov. 2021, 9, 100079. [Google Scholar] [CrossRef]
  164. Zhao, Q.; Xu, W.; Xing, L.; Lin, Z. Recombinant Production of Medium- to Large-Sized Peptides in Escherichia Coli Using a Cleavable Self-Aggregating Tag. Microb. Cell Fact. 2016, 15, 136. [Google Scholar] [CrossRef] [PubMed]
  165. Hou, W.; Zhang, X.; Liu, C.-F. Progress in Chemical Synthesis of Peptides and Proteins. Trans. Tianjin Univ. 2017, 23, 401–419. [Google Scholar] [CrossRef]
  166. Santos, L.; Oliveira, C.; Vasconcelos, B.M.; Vilela, D.; Melo, L.; Ambrósio, L.; da Silva, A.; Murback, L.; Kurissio, J.; Cavalcante, J.; et al. Good Management Practices of Venomous Snakes in Captivity to Produce Biological Venom-Based Medicines: Achieving Replicability and Contributing to Pharmaceutical Industry. J. Toxicol. Environ. Health Part B 2021, 24, 30–50. [Google Scholar] [CrossRef] [PubMed]
  167. Wang, L.; Wang, N.; Zhang, W.; Cheng, X.; Yan, Z.; Shao, G.; Wang, X.; Wang, R.; Fu, C. Therapeutic Peptides: Current Applications and Future Directions. Signal Transduct. Target. Ther. 2022, 7, 48. [Google Scholar] [CrossRef]
Figure 1. The process of detecting a noxious stimulus through nociceptors. Nociceptors feature a bifurcated projection: One branch extends as a peripheral axon, establishing connections with the skin and various organs, while the other branch projects toward the central nervous system (CNS). This intricate projection mechanism enables the transmission of information regarding the presence and intensity of the stimulus. The CNS then processes this information, orchestrating appropriate responses to potentially harmful stimuli.
Figure 1. The process of detecting a noxious stimulus through nociceptors. Nociceptors feature a bifurcated projection: One branch extends as a peripheral axon, establishing connections with the skin and various organs, while the other branch projects toward the central nervous system (CNS). This intricate projection mechanism enables the transmission of information regarding the presence and intensity of the stimulus. The CNS then processes this information, orchestrating appropriate responses to potentially harmful stimuli.
Pharmaceutics 15 02766 g001
Figure 2. Neurotransmitters involved in inflammatory pain sensation as all the major types of neurotransmitters (inflammatory mediators and non-inflammatory mediators), second messenger production, and others. Abbreviations: 5-HT: 5-hydroxytryptamine; 5-HT2A: 5-hydroxytryptamine type 2A receptor; 5-HT3: 5-hydroxytryptamine type 3 receptor; A1: adenosine type 1 receptor; A2: adenosine type 2 receptor; AC: adenylyl cyclase; AMPA-R: amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors; ASIC: acid-sensing ion channels; ATP: adenosine triphosphate; B1: bradykinin receptor type B1; B2: bradykinin receptor type B2; BK: bradykinin; CALCRL: calcitonin receptor-like receptor; cAMP: cyclic adenosine monophosphate; CB1: cannabinoid type 1 receptors; CB2: cannabinoid type 2 receptors; cGMP: cyclic guanosine monophosphate; CGRP: calcitonin gene-related peptide; Cl: chloride ion; DAG: diacylglycerol; DH: dorsal horn; DOR: δ-opioid receptors; EP: prostaglandin E2 receptor; EP1: prostaglandin E2 receptor type 1; EP2: prostaglandin E2 receptor type 2; EP3: prostaglandin E2 receptor type 3; EP4: prostaglandin E2 receptor type 4; EPSP: excitatory post-synaptic potentials; GABA: γ-aminobutyric acid; GABAA: γ-aminobutyric acid type A receptor; GABAB: γ-aminobutyric acid type B receptor; GlyR: glycine receptor; H1: histamine; IP: prostacyclin receptor; IP3: inositol triphosphate; K+: potassium ion; KOR: κ-opioid receptors; LTB4: leukotriene B4; LTB4-R1: leukotriene B4 type 1 receptor; LTB4-R2: leukotriene B4 type 2 receptor; Mg2+: magnesium ion; MOR: µ-opioid receptors; Na+: sodium ion; Nav: voltage-activated Na+ channels; NE: norepinephrine; NGF: nerve growth factor; NK1: neurokinin type 1 receptor; NK2: neurokinin type 2 receptor; NK3: neurokinin type 3 receptor; NKA: neurokinin A; NKB: neurokinin B; NMDA-R: N-methyl-D-aspartate receptors; NO: nitric oxide; P2X3: purino receptor; PAG: periaqueductal gray; PG: prostaglandins; PGE1: prostaglandin E1; PGE2: prostaglandin E2; PGI2: prostacyclin; PI3: phosphoinositide 3-kinase; PKA: protein kinase A; PKC: protein kinase C; PLC: phospholipase C; sGC: soluble guanylyl cyclase; SP: substance P; THC: tetrahydrocannabinol; VR1: vanilloid receptor for capsaicin; VSMC: vascular smooth muscle cell; α1: alpha 1-adrenoreceptor; α2: alpha 2-adrenoreceptor; β: beta-adrenoreceptor.
Figure 2. Neurotransmitters involved in inflammatory pain sensation as all the major types of neurotransmitters (inflammatory mediators and non-inflammatory mediators), second messenger production, and others. Abbreviations: 5-HT: 5-hydroxytryptamine; 5-HT2A: 5-hydroxytryptamine type 2A receptor; 5-HT3: 5-hydroxytryptamine type 3 receptor; A1: adenosine type 1 receptor; A2: adenosine type 2 receptor; AC: adenylyl cyclase; AMPA-R: amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors; ASIC: acid-sensing ion channels; ATP: adenosine triphosphate; B1: bradykinin receptor type B1; B2: bradykinin receptor type B2; BK: bradykinin; CALCRL: calcitonin receptor-like receptor; cAMP: cyclic adenosine monophosphate; CB1: cannabinoid type 1 receptors; CB2: cannabinoid type 2 receptors; cGMP: cyclic guanosine monophosphate; CGRP: calcitonin gene-related peptide; Cl: chloride ion; DAG: diacylglycerol; DH: dorsal horn; DOR: δ-opioid receptors; EP: prostaglandin E2 receptor; EP1: prostaglandin E2 receptor type 1; EP2: prostaglandin E2 receptor type 2; EP3: prostaglandin E2 receptor type 3; EP4: prostaglandin E2 receptor type 4; EPSP: excitatory post-synaptic potentials; GABA: γ-aminobutyric acid; GABAA: γ-aminobutyric acid type A receptor; GABAB: γ-aminobutyric acid type B receptor; GlyR: glycine receptor; H1: histamine; IP: prostacyclin receptor; IP3: inositol triphosphate; K+: potassium ion; KOR: κ-opioid receptors; LTB4: leukotriene B4; LTB4-R1: leukotriene B4 type 1 receptor; LTB4-R2: leukotriene B4 type 2 receptor; Mg2+: magnesium ion; MOR: µ-opioid receptors; Na+: sodium ion; Nav: voltage-activated Na+ channels; NE: norepinephrine; NGF: nerve growth factor; NK1: neurokinin type 1 receptor; NK2: neurokinin type 2 receptor; NK3: neurokinin type 3 receptor; NKA: neurokinin A; NKB: neurokinin B; NMDA-R: N-methyl-D-aspartate receptors; NO: nitric oxide; P2X3: purino receptor; PAG: periaqueductal gray; PG: prostaglandins; PGE1: prostaglandin E1; PGE2: prostaglandin E2; PGI2: prostacyclin; PI3: phosphoinositide 3-kinase; PKA: protein kinase A; PKC: protein kinase C; PLC: phospholipase C; sGC: soluble guanylyl cyclase; SP: substance P; THC: tetrahydrocannabinol; VR1: vanilloid receptor for capsaicin; VSMC: vascular smooth muscle cell; α1: alpha 1-adrenoreceptor; α2: alpha 2-adrenoreceptor; β: beta-adrenoreceptor.
Pharmaceutics 15 02766 g002
Table 1. Snake-derived toxins able to inhibit pain.
Table 1. Snake-derived toxins able to inhibit pain.
Toxin/
Molecule
Species ProductionAdministrationMechanism of AnalgesiaModelRef.
µ-EPTX-Na1Naja atraPurified from venomIntraplantar (i.pl.) injectionNav1.8 channel inhibitorAcute inflammation models induced by formalin and acetic acid, chronic inflammation pain model induced by complete Freund’s adjuvant and partial nerve ligation-induced allodynia.[69]
Mambalgin-1Dendroaspis polylepis polylepisPurified from venomIntravenous (i.v.) and intratechal (i.t.) injectionAcid-sensing ion channel (ASIC) inhibitorsMotor behavior tests such as accelerated rotarod test and grip strength test. Pain and inflammation models induced by carrageenan, thermal and mechanical pain test by von Frey[68]
Mambalgin-3Dendroaspis polylepis polylepisPurified from venomIntravenous (i.v.), intrathecal (i.t.), and intraplantar (i.pl.) injectionAcid-sensing ion channel (ASIC) inhibitorsMotor behavior tests such as accelerated rotarod test and grip strength test. Pain and inflammation models induced by carrageenan, thermal and mechanical pain test by von Frey[68]
Cobra neurotoxinNaja naja atraPurified from venomIntraperitoneal (i.p.) injectionAdenosine receptor (A1 and A2A) pathway activationAcute pain model induced by hot plate and spinal cord injury[74]
NajanalgesinNaja naja atraPurified from venomIntrathecal (i.t.) injectionc-Jun N-terminal kinase (JNK) inhibitorNeuropathic pain induced by spinal nerve ligation[75]
Crotoxin (CTX)Crotalus durissus terrificusPurified from venomSubcutaneous injection (s.c.)Formyl Peptide, α2-Adrenergic and Muscarinic ReceptorsAcute and chronic phases of hypernociception induced by partial sciatic nerve ligation[70]
Crotoxin (CTX)Crotalus durissus terrificusPurified from venomSubcutaneous injection (s.c.)Analgesia dependent on formyl peptide, lipoxygenase and muscarinic receptorsPain on the MOG 35-55-induced experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis[71]
CrotalphineCrotalus durissus terrificusChemical synthesisOral administration (p.o.) TRPA1The ciguatoxin-induced cold hypersensitivity, and the bradykinin-induced and zymosan-induced mechanical hypersensitivity[73]
Cobratoxin
(CbTX)
Naja naja kaouthiaPurified from venomIntrathecal (i.t.) injectionα7 nicotinic acetylcholine receptor (nAChRs)Acute pain model induced by hot plate and tail-flick[76]
Table 2. Scorpion-derived toxins able to inhibit pain.
Table 2. Scorpion-derived toxins able to inhibit pain.
Toxin/
Molecule
Species ProductionAdministrationMechanism of AnalgesiaModelRef.
rAGAPButhus martensiiRecombinantInjection application location not specifiedCHis6-rAGAP and NHis6-rAGAPXenograft tumor mouse model[78]
AGAP W38GButhus martensiiPurified from venomIntraplantar (i.pl.) injectionNav1.7 and Nav1.8 channel inhibitorAn acute thermal pain model induced by a hot plate and an acute inflammation model induced by formalin[79]
BmK AGP-SYPU1Buthus martensiiRecombinantIntraperitoneal (i.p.) injectionArginine residue at position 58 Mouse-twisting pain model[85]
BmK AGAPButhus martensiiRecombinantIntrathecal (i.t.) injectionMAPK inhibitorAn acute inflammation model induced by formalin, a thermal pain model induced by hot plate, and mechanical allodynia[80]
BotAFButhus occitanus tunetanusPurified from venomIntraperitoneal (i.p.), intrathecal (i.t.) and intraplantar (i.pl.) injectionsIon channel inhibitorAn acute thermal pain model induced by a hot plate, a nociception model induced by a shutter-controlled lamp, and an inflammation model induced by formalin and acetic acid writhing[81]
BmK AGAPButhus martensiiPurified from venomIntraplantar (i.pl.) injectionKv1.3 channel and MAPK inhibitor An acute inflammation model induced by formalin[82]
BmK AGAPButhus martensiiRecombinantIntraperitoneal (i.p.) injectionMCF-7 and MDA-MB-231 migration inhibitorXenograft tumor mouse model[77]
Makatoxin-3
(MkTxs)
Buthus martensiiPurified from venomIntraperitoneal (i.p.) injectionNav1.7 inhibitorAcute nociception induced by formalin test and Freund’s adjuvant (CFA) induced mechanical pain model[86]
TsNTxPTityus serrulatus-Intraperitoneal (i.p.) injectionNav channel inhibitorAcute nociception induced by a water bath and neuropathic pain model induced by CCI model[83]
LeptucinHemiscorpius lepturusChemical synthesisIntraperitoneal (i.p.) injectionIon channel inhibitorAcute thermal pain model induced by hot plate and nociception model using tail flick test[84]
Table 6. Mollusk-derived toxins able to inhibit pain.
Table 6. Mollusk-derived toxins able to inhibit pain.
Toxin/
Molecule
Species ProductionAdministrationMechanism of AnalgesiaModelRef.
Im10A (conotoxin)Conus imperialisSynthesizedIntramuscular
injection (i.m.)
-Rat PNL model[131]
μ-Conotoxin TsIIIAConus tessulatusSynthesizedIntrathecal (i.t.) injectionInhibition of TTX (tetrodotoxin)-resistant sodium currents in DRG neuronsHot-plate model[132]
α-Conopeptide Eu1.6Conus eburneusSynthesizedIntramuscular
injection (i.m.) and intravenous (i.v.) injection
Inhibition of high voltage-activated N-type calcium channel currents (Cav2.2) in isolated mouse dorsal root ganglia (DRG) neuronsRat partial sciatic nerve injury (PNL model) and chronic constriction injury pain (CCI model) models[102]
ω-Conotoxins: MoVIA and MoVIBConus
Moncuri
Purified from
venom and synthesized
Intrathecal (i.t.) injectionInhibition of rat Cav2.2 channelsPNL-induced neuropathic pain[134]
α-Conotoxin Vc1.1 Variants - SynthesizedIntramuscular (im.i), injectionInhibition of G protein-coupled γ-aminobutyric acid type B receptors (GABABR) coupled Cav2.2 channels in rat DRG neuronsPNL and CCI models[135]
cVc1.1 and cVc1.1 analogues: [C2H, C8F]cVc1.1 and [N9W]cVc1.1 - SynthesizedIntra-colonic administrationReduction in the excitability of DRG neuronsChronic visceral hypersensitivity (CVH model)[142]
ω -Conotoxin MVIIA modified - SynthesizedIntravenously (i.v.) injection
and intranasally injection
-Hot-Plate model[144]
αO-Conotoxin GeXIVAConus generalisSynthesizedIntramuscular (im.i), injection-Oxaliplatin-induced neuropathic pain, cold and mechanical allodynia[137]
Partially purified conotoxins (C1-C7) of C. coronatus and C. frigidus (F1-F6).Conus coronatus and Conus frigidusPurified from
Venom
Intraperitoneal (i.p.)
injection
Only C2 had analgesic effects in both tested models; mechanism of analgesia: not studiedHot-plate model and formalin-induced pain[145]
BuIA conotoxin analoguesConus bullatusSynthesizedIntraperitoneal (i.p.)
injection
All the analogs showed the same analgesic activity of BuIA
Mechanism of analgesia: not studied
Hot-plate model and paclitaxel-induced neuropathic pain[138]
ω-conotoxin Bu8Conus bullatusSynthesizedIntraperitoneal (i.p.)
injection
Inhibition of Cav2.2Hot-plate model and analgesic activity to acute pain and inflammatory pain[146]
Conotoxin-Ac1//
Conotoxin-Ac1-O6P (variant)
Conus achatinusSynthesizedIntrathecal (i.t.) injectionInhibition of NR2B ion channelsHot-plate and tail-flick models[141]
α-Conotoxin Vc1.1 modifiedConus victoriaePurified from
venom and modified
Intramuscular (im.i), injectionActivation of GABABRs expressed in DRGSpinal nerve ligation-induced neuropathic pain (SNL model), mechanical allodynia[136]
α4/7-Conotoxin (Lv1d)Conus lividusSynthesizedIntrathecal (i.t.) injection (mouse hot plate model)
Intramedullary injection (formalin-induced pain model)
-Mouse hot-plate and formalin-induced pain models[147]
µ-Conotoxin S24aConus striatusSynthesizedIntrathecal (i.t.) injection-Mouse hot-plate and formalin-induced pain models[148]
ω-ConotoxinConus virgoPurified from
venom and modified
Intraperitoneal (i.p.)
injection
-Central analgesic assay (tail immersion test) and Peripheral analgesic assay (acetic acid-induced writhing test)[149]
α-CTx RgIA analogues
(RgIA-5524)
- SynthesizedSubcutaneous
(i.s.) injection
Inhibition of α9-containing nicotinic acetylcholine receptors (nAChRs)Oxaliplatin-induced neuropathic pain[143]
α-Conotoxin RgIA analogues (RgIA-5474) - SynthesizedSubcutaneous
(i.s.) injection
-Oxaliplatin-induced neuropathic pain, cold allodynia[139]
ω–Conotoxins: MVIIA, GVIA and CVIF - Obtained from Alomone
Labs (Jerusalem, Israel).
Intraplantar (i.pl.). injection-Post-surgical pain (PSP model) and cisplatin-induced pain neuropathy and oxaliplatin-induced neuropathic pain[140]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pereira, A.F.M.; Cavalcante, J.S.; Angstmam, D.G.; Almeida, C.; Soares, G.S.; Pucca, M.B.; Ferreira Junior, R.S. Unveiling the Pain Relief Potential: Harnessing Analgesic Peptides from Animal Venoms. Pharmaceutics 2023, 15, 2766. https://doi.org/10.3390/pharmaceutics15122766

AMA Style

Pereira AFM, Cavalcante JS, Angstmam DG, Almeida C, Soares GS, Pucca MB, Ferreira Junior RS. Unveiling the Pain Relief Potential: Harnessing Analgesic Peptides from Animal Venoms. Pharmaceutics. 2023; 15(12):2766. https://doi.org/10.3390/pharmaceutics15122766

Chicago/Turabian Style

Pereira, Ana Flávia Marques, Joeliton S. Cavalcante, Davi Gomes Angstmam, Cayo Almeida, Gean S. Soares, Manuela B. Pucca, and Rui Seabra Ferreira Junior. 2023. "Unveiling the Pain Relief Potential: Harnessing Analgesic Peptides from Animal Venoms" Pharmaceutics 15, no. 12: 2766. https://doi.org/10.3390/pharmaceutics15122766

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop