Skip to main content
British Journal of Pharmacology logoLink to British Journal of Pharmacology
. 2016 Apr 22;173(11):1719–1727. doi: 10.1111/bph.13486

Possible role for anisodamine in organophosphate poisoning

Arik Eisenkraft 1,2,3,, Avshalom Falk 1
PMCID: PMC4867748  PMID: 27010563

Abstract

In cases of organophosphate poisoning, patients are treated with a combination of antidotes. In addition to these poison‐directed antidotes, patients may require extra oxygen and artificial ventilation; other modalities may also be needed due to the wide range of toxic effects. Anisodamine is a belladonna alkaloid, and like other drugs from this family is non subtype‐selective muscarinic, and a nicotinic cholinoceptor antagonist, which has been employed in traditional Chinese medicine. As a muscarinic antagonist, it displays similar pharmacological effects to atropine and scopolamine. However, anisodamine is not only less potent than atropine and scopolamine but also less toxic. Current in vitro and animal model studies have demonstrated that anisodamine has protective effects in a variety of diseases. Organophosphate poisoning involves not only the central and peripheral nervous systems, but also the cardiac and respiratory systems, as well as activation of inflammatory processes and oxidative stress. Therefore, the anticholinergic and additional activities of anisodamine appear to be relevant and justify its consideration as an addition to the existing remedies. However, more research is needed, as at present data on the role of anisodamine in the management of organophosphate poisoning are limited. Here, we review the beneficial effects of anisodamine on processes relevant to organophosphate poisoning.


Abbreviations

α7nAChR

α7 nicotinic ACh receptor

ALI

acute lung injury

ARDS

acute respiratory distress syndrome

mAChRs

muscarinic ACh receptors

nAChRs

nicotinic ACh receptors

OP

organophosphate

PMVEC

pulmonary microvascular endothelial cells

QNB

3‐quinuclidine‐benzylate

Th1/2

T‐helper cells 1 and 2

TSST‐1/2

toxic shock syndrome toxin type‐1/2

Tables of Links

These Tables list key protein targets and ligands in this article which are hyperlinked to corresponding entries in http://www.guidetopharmacology.org, the common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY (Pawson et al., 2014) and are permanently archived in the Concise Guide to PHARMACOLOGY 2015/16 (a,b,cAlexander et al., 2015a, 2015b, 2015c).

Introduction

In cases of organophosphate (OP) poisoning, it is accepted that patients should be treated with a combination of an anti‐cholinergic drug, usually atropine, an oxime as a cholinesterase reactivator, and if the patient develops convulsions and seizures, a benzodiazepine such as diazepam or midazolam is added (Eddleston et al., 2006, 2008; Rosman et al., 2014). Together with the abovementioned drugs, it is important to use supplemental oxygen and ventilate the casualty if needed (Markel et al., 2008; Sawyer et al., 2012). However, too high a dose of atropine may lead to excessive anticholinergic toxicity, which may even be fatal (Eddleston et al., 2004; Sidell et al., 2008; Wang et al., 2014b).

Anisodamine is a belladonna alkaloid, and like other drugs from this family is a non‐subtype‐selective muscarinic, and also a nicotinic cholinoceptor antagonist (Zhao et al., 1993). It has been employed in traditional Chinese medicine for many ailments, mainly to improve the microcirculation in states of shock, and also in organophosphate poisoning (Poupko et al., 2007; Wang et al., 2014a, 2014b). However, the clinical toxicological information available on anisodamine in western countries is limited. As a muscarinic antagonist, it displays similar pharmacological effects to atropine and scopolamine, including inhibition of secretion (e.g. salivation, diaphoresis and respiratory secretions), bronchodilation, inhibition of gastrointestinal motility and changes in cardiovascular function (Anon. 1975; Zang et al., 1987; Guo et al., 1992a; Pan and Han, 2000; Wang et al., 2003a; Pan and Han, 2004). Several studies have shown that anisodamine is at least one order of magnitude less potent than atropine and scopolamine (Anon. 1975; Zang et al., 1987; Guo et al., 1992a; Pan and Han, 2000; Wang et al., 2003a; Poupko et al., 2007). Relatively little clinical information is available on its human toxicity. Based on animal studies, it seems to be less toxic than atropine and scopolamine (Poupko et al., 2007). The usual treatment dose in humans is 10 mg·kg−1, yet doses as high as 500 mg·kg−1·day−1 did not produce any serious adverse effects (Li et al., 1999). The lower potency of anisodamine and the finding that it is less likely to impair learning and memory when compared with atropine are probably related to its decreased lipid solubility and limited blood–brain barrier permeability (Zhang, 2002). Current in vitro and animal model studies have demonstrated that anisodamine has beneficial effects in a variety of disease conditions of different aetiologies and in various organs. These include cardiac arrhythmias (Chen and Gu, 1988; Yang et al., 1991), acute lung injury (Zhang et al., 2005), asthma (Xu et al., 2011), arthritis (Zhou et al., 2014) and septic shock (Zhao et al., 2011). It was found to be protective at cellular, organ system and animal levels in inflammation (Liu et al., 2009; Xu et al., 2011; You et al., 2014), oxidative stress (Luo et al., 1992; Esberg and Ren, 2004; Liu et al., 2013), coagulopathy (Xiu et al., 1982; Ruan et al., 2001, 2002) and molecular signalling pathways (Norby and Ren, 2002; Xing et al., 2015).

OP poisoning, either by pesticides or chemical warfare nerve agents, is characterized by a complex toxidrome involving more than one body system – mainly the central and peripheral nervous systems and the cardiorespiratory system (Markel et al., 2008; Sidell et al., 2008; Hulse et al., 2014). In fact, regulation of all organ systems involves ACh and its receptors (Wessler and Kirkpatrick, 2008). Furthermore, the pathophysiology of OP poisoning involves, in addition to cholinergic effects, inflammatory processes and oxidative stress in the affected organs (Collombet, 2011; Banks and Lein, 2012). In this manuscript, the effects of anisodamine on these processes will be reviewed in order to present the broad spectrum of potential beneficial effects relevant to the management of OP poisoning, starting with the studies on the use of anisodamine in the management of OP poisoning in humans and in counteracting the toxicity of related pharmacological agents in animals.

Anisodamine as an antidote in OP poisoning

Anisodamine has long been considered as an antidote in OP poisoning in China and Tibet (Poupko et al., 2007; Wang et al., 2014a, 2014b). As a muscarinic antagonist, it has similar effects to those of atropine and scopolamine, including reducing salivation, lacrimation and sweat, diminishing gastrointestinal motility, mydriasis, bronchoconstriction and cardiovascular changes characteristic of the toxicity of muscarinic agonists (Poupko et al., 2007; Wang et al., 2014a, 2014b). It is less potent than scopolamine at inducing central effects (Poupko et al., 2007; Zhang et al., 2008, 2009), and in septic shock, it was reported to show fewer adverse effects than atropine (Li et al., 1999). In a neuropsychopharmacological study in mice, anisodamine did not cause spatial cognitive deficits and even improved cognition at repeated high doses, based on Morris water maze test, and did not depress LTP recordings in rats, while scopolamine has detrimental effects on both activities (Zhang et al., 2008). This difference in in vivo effects was attributed by the authors to the poor blood–brain barrier permeability of anisodamine compared with scopolamine, with estimated octanol–water partition coefficient (logP) values of 0.25 and 0.76, respectively (Zhang et al., 2008). These differences could also be attributed in part to its lower binding affinity to brain mAChRs compared to scopolamine; this was measured in mouse brain homogenates by competitive displacement of [3H]‐quinuclidinyl benzilate (QNB), showing IC50 values of 72.0 ± 1.7 and 0.18 ± 0.02 μM for anisodamine and scopolamine, respectively (Zhang et al., 2008). This is important in case of blood–brain barrier dysfunction, which was shown to occur in patients with severe CNS toxicity due to nerve agents or OP pesticides (Abdel‐Rahman et al., 2002; Song et al., 2004; Testylier et al., 2007).

The potential of anisodamine in ameliorating systemic cholinergic toxicity is evident from studies in a mouse model of cognitive impairment, where a cholinergic deficiency was induced by a high dose of scopolamine to mimic human Alzheimer's disease (Wang et al., 2003a; Zhang et al., 2009). In an earlier study, this cognitive impairment could be overcome by administration of the muscarinic agonist pilocarpine (20 or 40 mg·kg−1 i.p. twice daily), but this was accompanied by typical muscarinic side effects of salivation, enhanced bowel movement and bradycardia. However, these were ameliorated by 20 mg·kg−1 anisodamine, with similar cognitive improvements (Wang et al., 2003a).

The same research group showed later that administration of anisodamine to mice injected with scopolamine and treated with the AChE inhibitor rivastigmine counteracted the muscarinic and neuromuscular nicotinic‐mediated adverse effects of rivastigmine, which included hypersalivation, hyperperistalsis and muscle cramps, while facilitating cognitive amelioration (Zhang et al., 2009). This may indicate a potential role for the combination of both drugs as a prophylactic treatment against OP poisoning, nerve agents in particular. We have previously shown that rivastigmine can act as a prophylactic drug in nerve agent poisoning in animal models. However, in a human safety study (phase I), we observed that it had adverse effects, which would prevent its use as a single compound for such an indication (Lavon et al., 2015). The results presented here indicate that the combination of anisodamine and rivastigmine could serve as a future prophylactic mixture. This is yet to be studied.

As a cholinergic antagonist, anisodamine can inhibit cholinergic hyperexcitability, reduce hypersecretion, stimulate the respiratory centre, relax the spasm of smooth muscle, accelerate the heart rate and abolish other muscarinic symptoms (Wang et al., 2014a). Wang et al. (2014a) described a patient with severe OP poisoning. She was admitted in a moderate clinical condition, and despite receiving the accepted treatment protocol, within 4 h from admission, her clinical condition had deteriorated, and she was intubated and ventilated. Twenty‐two hours after being poisoned, she received a total of 960 mg atropine i.v., 3.25 g pralidoxime i.v., and her serum AChE activity had changed from 59% on admission to 72% of the normal activity level, but she still needed mechanical ventilation. At this stage, treatment with anisodamine was initiated. Four hours later, 26 h after being poisoned, a total of 480 mg anisodamine and 0.5 g pralidoxime was given i.v., AChE activity was restored to 75% of normal range, and her blood gases returned to normal. On the following day, she received four more doses of anisodamine, 20 mg each. Her clinical condition gradually improved, 5 days post exposure she was extubated, and on day 9, she was released from hospital. Anisodamine was given for 3 days. This case report shows that anisodamine had a better clinical outcome than high and repeated doses of atropine (Wang et al., 2014a).

The same group of researchers performed a retrospective analysis of 64 OP‐poisoned patients (Wang et al., 2014b). All of them received the routine, accepted antidote treatment, consisting of repeated atropine and pralidoxime doses, yet in all, atropinization was not achieved after 12 h, following high doses of atropine. Thirty‐six patients continued with the original atropine treatment protocol, while 28 started an anisodamine treatment protocol at this stage. In the atropine group, time to atropinization was 29 ± 7.0 h, compared with 24.3 ± 4.3 h in the anisodamine group (P < 0.05). Hospital stay in the anisodamine group was 5.3 ± 2.5 days compared with 6.9 ± 2.3 days in the atropine‐only group (P < 0.05). This study indicates that anisodamine can shorten the time to atropinization and reduce the hospital stay of OP‐poisoned patients. The time to reach complete atropinization is critical, as a delay in atropinization may increase the likelyhood of the establishment of irreversible damage. Furthermore, there are still concerns regarding the toxicity of the administration of high doses of atropine (Eddleston et al., 2004). A comparison of existing atropine treatment protocols in OP poisoning has shown that the protocols are poorly evidence‐based, highly variable between published sources, and most of them are too slow to achieve the desired atropinization dose in time (Eddleston et al., 2004). A follow‐up study has shown some change towards faster dosing protocols, but the variability still exists (Connors et al., 2014), which may be one of the reasons why atropinization is difficult to attain in some severely poisoned patients (Kaur et al., 2014; Iyer et al., 2015). Recent reviews covering novel and alternative ways for treating OP poisoning (Kaur et al., 2014; Iyer et al., 2015) have supported the Wang et al. (2014a, 2014b proposal that anisodamine can be used as a safe adjunct or substitute to atropine, for cases where atropinization is difficult to achieve.

Anti‐oxidant and membrane effects of anisodamine

Anisodamine is known to possess an anti‐oxidant action and can protect against free radical‐induced cellular damage. It has been shown to interact and disrupt liposomes similar to the cell's membrane lipid bilayer, by increasing their fluidity (Hwang et al., 1983). Anisodamine was found to interact with membrane acidic phospholipids, which play an important role in neural cells. This may explain its ability to protect nerve cells (Wang et al., 1993). Luo et al. (1992) have shown that anisodamine exerted a protective effect in cultured bovine pulmonary endothelial cells from injury induced by oxygen free radicals (Luo et al., 1992). The authors noted that there was no evidence of any scavenging effects of anisodamine and, therefore, these findings also support the hypothesis that anisodamine is a membrane stabilizing compound (Luo et al., 1992). In a swine model of cardiac arrest, Liu et al. (2013) found that anisodamine‐treated animals had significantly lower plasma levels of malondialdehyde, elevated levels of superoxide dismutase and ATP and lower levels of mitochondrial ROS, when compared with controls and an adrenaline‐treated group (Liu et al., 2013). Mitochondrial injury, observed using transmission electron microscopy, was milder than in the other two groups. They concluded that anisodamine alleviates mitochondrial oxidative‐induced injury in the myocardium by regulating the metabolism of ROS.

These properties of anisodamine, together with its anticholinergic effects, may add to its value as a therapy for OP poisoning, as an antioxidant treatment has already been shown to be beneficial in such cases (Elsinghorst et al., 2013).

Role of anisodamine in the regulation of immune and inflammatory responses

There is growing evidence that the cholinergic anti‐inflammatory pathway has an important role in regulating both immune and inflammatory responses. Activation of mAChRs and nAChRs stimulate and inhibit the immune and inflammatory responses, respectively (Razani‐Boroujerdi et al., 2008). The cholinergic anti‐inflammatory pathway denotes the interaction between the nervous and the immune systems, specifically between the efferent vagus nerve and peripheral α7 nicotinic ACh receptors (α7nAChRs) expressed on monocytes and macrophages (Pavlov et al., 2003; Rosas‐Ballina and Tracey, 2009). Through vagal release of ACh, the nervous system can significantly inhibit the ability of macrophages to release TNF‐α, thus attenuating the systemic inflammatory responses (Wang et al., 2003b; Rosas‐Ballina and Tracey, 2009). In the CNS, signalling through α7nAChRs in microglia has an anti‐inflammatory effect, similar to the effect found in macrophages (Shytle et al., 2004). As a muscarinic receptor antagonist, anisodamine may lead to increased ACh‐mediated activation of α7nAChR, together with muscarinic receptor inhibition, which in turn activates the cholinergic anti‐inflammatory pathway and controls cytokine production (Liu et al., 2009; Zhao et al., 2011). Anisodamine was shown to significantly reduce TNF‐α and IL‐1β production in both in vitro and in vivo models of LPS‐induced inflammation and shock (Liu et al., 2009). The proposed mechanism was increased activation of α7nAChR by endogenous ACh, diverted from the anisodamine‐inhibited muscarinic AChR (Liu et al., 2009). A later study from this group suggested that anisodamine acts synergistically with IL‐10 to up‐regulate the expression of α7nAChR (Li et al., 2011). However, as not all of the effects of LPS were abolished by a selective α7nAChR antagonist, other mechanisms besides the cholinergic anti‐inflammatory pathway, such as antagonism of adrenoceptors by large doses, could not be ruled out (Liu et al., 2009). Moreover, the ability of atropine to protect mice from LPS‐induced lethality was found to be independent of IL‐10 (Fuentes et al., 2008), thus pointing to more than one mechanism of protection by cholinolytics. The combined treatment of anisodamine with the peripheral reversible cholinesterase inhibitor neostigmine was more effective than each drug alone in protecting rats from LPS lethality and in reducting blood levels of TNF‐α and IL‐1β and from haemorrhagic shock in beagle dogs (Sun et al., 2012). This combination allowed both drugs to be used at a lower dose, so reducing the adverse effects of neostigmine, and was also found to be protective in other models of severe or debilitating disease involving inflammation, such as obstructive jaundice, collagen‐induced arthritis and ischaemic stroke (Li et al., 2014; Zhou et al., 2014; Qian et al., 2015).

Anisodamine was found to protect animals from toxic shock syndrome toxin‐1 (TSST‐1) induced by Staphylococcus aureus (Nakagawa et al., 2005) and from Shiga toxin type 2 (Zhang et al., 2001). In both studies, the protective effect involved the down‐regulation of pro‐inflammatory cytokines like TNF‐α, and in the case of TSST‐1, the effect was mediated by inhibition of NF‐κB activation (Nakagawa et al., 2005).

In a model of brief forebrain ischaemia–reperfusion in gerbils, a condition involving inflammation, treatment with anisodamine resulted in a large number of viable neurons in the CA1 subfield in the hippocampus, compared with the untreated group (Chen and Zeng, 2000). The researchers also performed the open‐field test as a sensitive tool for the assessment of memory deficits resulting from hippocampal cell loss. Anisodamine reduced the open‐field habituation impairment in the ischaemic animals. Moreover, anisodamine appeared to inhibit the production of hydroxyl radicals (Chen and Zeng, 2000). Inflammation and oxidative stress are hallmarks in OP‐induced CNS pathology (Collombet, 2011; Banks and Lein, 2012; Iyer et al., 2015). The anti‐inflammatory, antioxidant and cell‐membrane membrane‐protective properties of anisodamine are consistent with its neuroprotective role in this case.

Pulmonary effects

The most imminent threat in OP poisoning results from pulmonary insufficiency. This is the result of hypersecretion, respiratory muscle paralysis and bronchoconstriction (Eddleston et al., 2006; Markel et al., 2008; Sidell et al., 2008; Hulse et al., 2014; Rosman et al., 2014). Moreover, damage to the respiratory centres of the brain adds to the respiratory compromise, as it is well known that severe brain injury, including in cases of OP poisoning, is often associated with pulmonary insufficiency (Eddleston et al., 2006; Carey et al., 2013). The immediate local pulmonary effects of OPs – bronchoconstriction and hypersecretion – are reversed by atropine and scopolamine (Nambiar et al., 2007; Perkins et al., 2011a, 2011b), indicating that a local excess of ACh due to inhibition of cholinesterases is involved in this effect (Graham et al., 2006). Similarly, in chronic respiratory diseases like asthma and chronic obstructive pulmonary disease, respiratory symptoms are mediated by ACh (Gosens et al., 2006) and modulated both experimentally and clinically by anticholinergic drugs (Moulton and Fryer, 2011).

As a non‐selective muscarinic antagonist, anisodamine was found to alleviate acute lung injury (ALI) induced by a variety of causes in animal models. Both clinical and experimental lung injuries can be induced by direct insults like aspiration, pneumonia or inhalation of toxic gases, or indirectly by trauma, sepsis and systemic inflammatory response in general (Perl et al., 2011; Matthay et al., 2012). The effect of anisodamine in clinical secondary acute lung injury was demonstrated by Huang et al. (2002), who showed that patients with brain damage treated with anisodamine in addition to the standard therapy had improved pulmonary function and increased alveolar and arterial oxygenation, when compared with patients receiving standard care alone (Huang et al., 2002). The authors linked this effect to anisodamine‐induced inhibition of TNF‐α and IL‐6, which are regarded as mediators of such an injury.

The LPS of gram‐negative bacteria can lead to ALI through both direct and indirect effects (Menezes et al., 2005; Perl et al., 2011). This includes direct injury to endothelial cells and indirect injury due to activation of inflammatory cells. The injured tissue and activated inflammatory cells release pro‐inflammatory cytokines, including TNF‐α, IL‐1β, IL‐6 and IL‐17, specifically IL‐17A and IL‐17F, leading to additional leukocyte recruitment and damage in the inflamed airways and lungs (Ferretti et al., 2003; Miyamoto et al., 2003; Yang et al., 2008; Prause et al., 2009). The involvement of the cholinergic system in LPS‐induced lung injury was demonstrated directly by Xu et al. (2012). They showed that pulmonary dysfunction and inflammatory responses in mice treated intratracheally with LPS could be attenuated by either α7nAChR agonist or the non‐subtype selective muscarinic antagonist atropine or the specific M3 receptor antagonist 4‐DAMP but not specific M1 or M2 receptor agonists, thus showing the role of the M3 subtype in induction of lung injury in this model. Activation of pulmonary microvascular endothelial cells (PMVECs) is considered to be an early step in the inflammation process of ALI (Wu et al., 2008). You et al. (2014) performed an in vitro study in rat PMVECs and an in vivo study in a rat model of LPS‐induced ALI to test whether anisodamine has a modulatory effect and further to see if it involves IL‐17 expression. They showed that anisodamine reduced lung injury and the expression of IL‐17A and IL‐17F in PMVECs and in rats following exposure to LPS. Furthermore, the reduction in lung injury and IL‐17A/IL‐17F production in LPS‐treated animals were comparable with that of the known anti‐inflammatory drug methylprednisolone. Studies with anti‐inflammatory agents showed promising results in preclinical models and small‐scale clinical studies but did not provide conclusive evidence of efficacy in larger clinical trials, prompting further research into this important strategy (Matthay et al., 2012). Clinical studies with methylprednisolone showed improvement in clinical parameters but no conclusive evidence of reduced mortality (Meduri et al., 1998, 2002, 2007). The results of You et al. (2014) indicate a possible role for anisodamine in the treatment of sepsis‐induced ALI, prompting additional research to investigate this. Anisodamine was found to be protective in other animal models of indirect ALI. In a canine cardiopulmonary bypass model, anisodamine reduced lung injury by significantly reducing lung water content, pulmonary granulocyte sequestration and oxygen free‐radical release compared with untreated controls (Gu et al., 1991). Anisodamine also had a protective effect in lungs in a rabbit lung injury model following exposure to oleic acid (Deng et al., 1989).

The protective effect of anisodamine in injury mediated by inhalation of lung‐damaging toxic industrial chemicals was demonstrated after inhalation of the toxic industrial compound perfluoroisobutylene (PFIB), an electrophilic toxic compound similar to phosgene but with higher potency (Zhang et al., 2005). Lung injury by this compound results from both direct and indirect mechanisms. The direct injury is probably due to PFIB's electrophilic attack on nucleophilic groups of amino acids in the alveolar membranes (Zhang et al., 2005) and an impaired lung microcirculation contributes to the development of ALI. The major indirect effects result from neutrophil sequestration and accumulation, key factors involved in ALI in general (Grommes and Soehnlein, 2011; Perl et al., 2011; Matthay et al., 2012; Williams and Chambers, 2014) and together with other factors contribute to the injury evoked by PFIB and the resulting ALI (Zhang et al., 2005). Zhang et al. have shown that in a rat model of PFIB‐induced ALI, high doses of anisodamine (30 mg·kg−1) attenuated pulmonary oedema (decreased wet lung/body weight ratio, dry lung/body ration and total protein concentration) (Zhang et al., 2005). Similar effects and also a reduction in neutrophil recruitment, oxidative stress, ultrastructural changes in alveolar epithelial cells and decreased mortality, were observed in PFIB‐exposed mice treated with the more potent cholinolytic agent QNB (Zhang et al., 2005), suggesting a role for cholinolytic agents, including anisodamine, in the management of chemically‐induced lung injury.

In a mouse model of ovalbumin‐induced asthma, anisodamine was shown to significantly suppress the peribronchial and perivascular infiltration and accumulation of eosinophils in the airways, similar to the effects of atropine and dexamethasone (Xu et al., 2011). This indicates the probable involvement of muscarinic receptors in airway inflammation following eosinophil infiltration. This model of airway reactivity includes an increase in the contractile responses to ACh. As with dexamethasone, this increase was significantly inhibited by anisodamine. Anisodamine also attenuated airway epithelial hyperplasia and thickening of the basement membrane. Because anisodamine prevented bronchial contractile responses only following ovalbumin challenge, this may indicate that ACh affects inflamed airway smooth muscle. ACh is known to promote the release of cytokines from epithelial and smooth muscle cells during inflammation, which in turn attract inflammatory cells to the airways and induce remodelling of the tissue (Gosens et al., 2006; Xu et al., 2011; Kolahian and Gosens, 2012). In this study, anisodamine had a similar effect as atropine in that it attenuated eosinophilic airway inflammation and affected the balance of Th1/Th2 lymphocytes, suppressing the Th2‐mediated immune response and enhancing the Th1‐mediated immune response (down‐regulation of Th2‐associated IL‐4 and up‐regulation of Th1‐associated IFNγ, respectively) (Xu et al., 2011). This suggests that ACh is involved in these immune inflammatory responses, as also shown by Wen et al. (2006). More published data indicate that the cholinergic system has a role in immune responses and inflammation and because muscarinic receptors are involved in the regulation of Th1/Th2 (Wen et al., 2006; Razani‐Boroujerdi et al., 2008; Qian et al., 2011; Hwang et al., 2013), anisodamine and other muscarinic antagonists may act as important modulators of inflammation through their influence on non‐neural cholinergic systems. The immune‐modulatory effects of anisodamine might be mediated by affecting the balance of muscarinic and nicotinic receptor activation.

The inhibition of inflammatory processes and resulting amelioration of lung injury by α7nAChR agonists was shown in several models of ALI, demonstrating the role of cholinergic signalling through this receptor in the control of pulmonary inflammation (Wu et al., 2014). Taken together, these recent findings and the beneficial effects of anisodamine through α7nAChR in models of septic shock endorse the potential of anisodamine in the therapy of acute injury and chronic pulmonary diseases.

Another alternative mechanism is that by inhibiting the M2 inhibitory ACh receptors on cholinergic nerve terminals, anisodamine contributes to the release of ACh in the cholinergic anti‐inflammatory pathway (Rosas‐Ballina and Tracey, 2009). A reduction in M2 receptors has been shown to contribute to a hypercholinergic immunosuppressive state, both in the brain and in the periphery (Allon et al., 2011).

Early in acute respiratory distress syndrome (ARDS), activated neutrophils are sequestered in the lungs and produce ROS and other products leading to lung tissue damage (Grommes and Soehnlein, 2011; Williams and Chambers, 2014). This is an important mechanism in the initiation of ARDS, and anisodamine may help in preventing any further deterioration. Anisodamine seems to possess both anti‐oxidative and superoxide scavenging activities and was used in both experimental and clinical cases of ARDS (Yu et al., 1985; Luo et al., 1987, 1992). As mentioned earlier, anisodamine protects tissues from oxygen free‐radical‐induced injury probably through its membrane‐stabilizing properties (Luo et al., 1992).

In an early study on systemic and pulmonary circulations in rabbits, anisodamine was found to dilate both pulmonary and systemic vessels, even though the effect was defined as relatively weak (Fan et al., 1986).

Cardiovascular effects

Current evidence shows that anisodamine depresses cardiac conduction and also has a dose‐dependent anti‐arrhythmic effect (Chen and Gu, 1988; Yang et al., 1991). Anisodamine inhibited cardiomyocyte contractility and the transient rise in intracellular Ca2 + concentration and also depressed cardiomyocyte contraction invoked by the muscarinic agonist carbachol (Norby and Ren, 2002). Anisodamine inhibited the uptake of Ca2 + by the skeletal muscle sarcoplasmic reticulum Ca2 + ATPase, possibly by inducing a structural modification of the transmembrane domain of the protein, which is the site of Ca2 + binding (Pang and Chen, 2004). This finding may provide an explanation for the modulating effect of anisodamine on cardiac function (Poupko et al., 2007). Arrhythmias may occur as delayed effects of OP poisoning, and these may be fatal (Allon et al., 2005; Bar‐Meir et al., 2007). In rats surviving sarin poisoning (exposure to ~LCt50), an acute and temporary QT prolongation was observed initially, followed by a long‐lasting increased sensitivity to adrenaline‐induced arrhythmias (Allon et al., 2005, 2011). Anisodamine was shown to decrease the adrenaline‐induced ventricular tachycardia in rabbits (Chen and Gu, 1988; Poupko et al., 2007). These findings warrant further studies on the role of anisodamine in controlling OP‐related arrhythmias.

Other direct protective effects of anisodamine on the heart stem from its antioxidant properties. Protection of the myocardium and myocardial mitochondria from oxidative damage in ischaemia–reperfusion was discussed above (Liu et al., 2013). Anisodamine attenuated the inhibition of cardiomyocyte contractility mediated by oxidative stress, invoked by the peroxide radical donor pyrogallol (Esberg and Ren, 2004). The protective effect may not result from oxygen radical scavenging but from alleviation of the oxidative stress‐induced Ca2 + overload by a direct effect on the sarcoplasmic reticulum (Esberg and Ren, 2004). In another study, anisodamine reduced cardiomyocyte apoptosis in a rat model of ischaemia–reperfusion (Xing et al., 2015).

Anisodamine improves blood flow in the microcirculation through various mechanisms, including (i) evoking vasodilatation by antagonizing ACh‐induced prejunctional inhibition of adrenergic transmission, antagonizing ACh‐induced endothelium‐dependent relaxation and α1‐adrenoceptor blockade (Varma and Yue, 1986; Guo et al., 1992b, 1992c, 1993); (ii) both direct and indirect effects on the heart (increased contractility and increased venous return and preload) (Yao et al., 1984); and (iii) an anti‐thrombotic and fibrinolytic effects, in which it inhibits platelet aggregation both in vitro, in vivo and in humans. The exact mechanism is still unclear but seems to include inhibition of thromboxane synthesis, inhibiting endothelial cell activation, exerting anti‐proteolytic activity and suppressing TNF‐α‐induced tissue factor, which is needed for thrombin activity (Xiu et al., 1982; Su et al., 1984; Xiao et al., 1988; Ruan et al., 2001; Zhang et al., 2001; Ruan et al., 2002). This may also explain its efficacy in cases of shock, coronary artery disease, thrombophlebitis and disseminated intravascular coagulation.

Conclusions

Taken together, it seems that anisodamine may exert its positive effects in multiple pathways, leading to an improvement in the clinical condition of OP‐poisoned patients faster than atropine. Although the number of publications on the role of anisodamine in the management of OP poisoning is small, the evidence from other research and clinical areas point strongly to its potential role in the management of OP poisoning. These include, in addition to cholinolytic activity per se, anti‐inflammatory, antioxidant, antiarrhythmic, anticoagulant and haemodynamic beneficial activities. Of these, the most prominent studies cited are in the alleviation of acute lung injury and cardiovascular effects, being the most serious and life‐threatening complications of peripheral OP toxicity, and requiring special attention. Also noted is the lower toxicity of anisodamine compared with that of atropine, and this may be important in cases when atropine is inefficient or contraindicated. The efficacy of anisodamine in the treatment of OP poisoning, either alone or in combination with other treatment modalities, should be further evaluated, both in the experimental and the clinical settings. Basic research on the mechanism of action of anisodamine with reference to other naturally derived and synthetic cholinolytic drugs is essential to evaluate and better define its role in the management of OP poisoning. These should include additional pharmacokinetic and receptor binding studies to identify the specific receptors and pathways involved in its pharmacological effects, identification of cholinergic and non‐cholinergic mechanisms in neural, pulmonary and cardiovascular effects of anisodamine and their integrative assessment by physiologically based pharmacokinetic/pharmacodynamic (PB/PK/PD) modelling.

Conflict of interest

The authors declare no conflicts of interest.

Eisenkraft, A. , and Falk, A. (2016) Possible role for anisodamine in organophosphate poisoning. British Journal of Pharmacology, 173: 1719–1727. doi: 10.1111/bph.13486.

References

  1. Abdel‐Rahman A, Shetty AK, Abou‐Donia MB (2002). Acute exposure to sarin increases blood brain barrier permeability and induces neuropathological changes in the rat brai: dose–response relationships. Neuroscience 113: 721–741. [DOI] [PubMed] [Google Scholar]
  2. Alexander SPH, Davenport AP, Kelly E, Marrion N, Peters JA, Benson HE et al. (2015a). The Concise Guide to PHARMACOLOGY 2015/16: G protein‐coupled receptors. Br J Pharmacol 172: 5744–5869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Alexander SPH, Peters JA, Kelly E, Marrion N, Benson HE, Faccenda E et al. (2015b). The Concise Guide to PHARMACOLOGY 2015/16: Ligand‐gated ion channels. Br J Pharmacol 172: 5870–5903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Alexander SPH, Fabbro D, Kelly E, Marrion N, Peters JA, Benson HE et al. (2015c). The Concise Guide to PHARMACOLOGY 2015/16: Enzymes. Br J Pharmacol 172: 6024–6109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Allon N, Rabinovits I, Manistersky E, Weissman BA, Grauer E (2005). Acute and long‐lasting cardiac changes following a single whole body exposure to sarin vapor in rats. Toxicol Sci 87: 385–390. [DOI] [PubMed] [Google Scholar]
  6. Allon N, Chapman S, Egoz I, Rabinovitz I, Kapon J, Weissman BA et al. (2011). Deterioration in brain and heart functions following a single sub‐lethal (0.8 LCt50) inhalation exposure of rats to sarin vapor: a putative mechanism of the long term toxicity. Toxicol Appl Pharmacol 253: 31–37. [DOI] [PubMed] [Google Scholar]
  7. Anonymous (1975). Pharmacologic effects of anisodamine. Chin Med J (Engl) 1: 133–138. [PubMed] [Google Scholar]
  8. Banks CN, Lein PJ (2012). A review of experimental evidence linking neurotoxic organophosphorus compounds and inflammation. Neurotoxicology 33: 575–584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Bar‐Meir E, Schein O, Eisenkraft A, Rubinshtein R, Grubstein E, Militianu A et al. (2007). Guideleines for treating cardiac manifestations of organophosphates poisoning with special emphasis on long QT and Torsade des Pointes. Crit Rev Toxicol 37: 279–285. [DOI] [PubMed] [Google Scholar]
  10. Carey JL, Dunn C, Gaspari RJ (2013). Central respiratory failure during acute organophosphate poisoning. Respir Physiol Neurobiol 189: 403–410. [DOI] [PubMed] [Google Scholar]
  11. Chen JM, Gu SL (1988). Anti‐arrhythmic effects of anisodamine. Yao Xue Xue Bao 23: 857–859. [PubMed] [Google Scholar]
  12. Chen Q, Zeng Y (2000). Anisodamine protects against neuronal death following cerebral ischemia in gerbils. Chin Med J (Engl) 113: 636–639. [PubMed] [Google Scholar]
  13. Collombet JM (2011). Nerve agent intoxication: recent neuropathophysiological findings and subsequent impact on medical management prospects. Toxicol Appl Pharmacol 255: 229–241. [DOI] [PubMed] [Google Scholar]
  14. Connors NJ, Harnett ZH, Hoffman RS (2014). Comparison of current recommended regimes of atropinization in organophosphate poisoning. J Med Toxicol 10: 143–147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Deng GH, Luo ZY, Yin D, Luo H (1989). Protective effects of anisodamine on lung injury induced by oleic acid in rabbits. Bull Hunan Med Coll 14: 339–342. [Google Scholar]
  16. Eddleston M, Buckley NA, Checketts H, Senarathna L, Mohamed F, Sheriff MH et al. (2004). Speed of initial atropinization in significant organophosphorus pesticide poisoning – a systematic comparison of recommended regimes. J Toxicol Clin Toxicol 42: 865–875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Eddleston M, Mohamed F, Davies JOJ, Eyer P, Worek F, Sheriff MJR et al. (2006). Respiratory failure in acute organophosphorous pesticide self‐poisoning. Q J Med 99: 513–522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Eddleston M, Buckley NA, Eyer P, Dawson AH (2008). Management of acute organophosphate pesticide poisoning. Lancet 371: 597–607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Elsinghorst PW, Worek F, Thiermann H, Wille T (2013). Drug development for the management of organophosphorus poisoning. Expert Opin Drug Discov 8: 1467–1477. [DOI] [PubMed] [Google Scholar]
  20. Esberg LB, Ren J (2004). The oxygen radical generator pyrogallol impairs cardiomyocyte contractile function via a superoxide and p38 MAP kinase‐dependent pathway: protection by anisodamine and tetramethylpyrazine. Cardiovasc Toxicol 4: 375–384. [DOI] [PubMed] [Google Scholar]
  21. Fan YL, Yang L, Wang XQ, Sun HL, Sun YW, Hu F et al. (1986). Effects of anisodamine and scopolamine on pulmonary circulation in rabbits. Zhongguo Yao Li Xue Bao 7: 117–121. [PubMed] [Google Scholar]
  22. Ferretti S, Bonneau O, Dubois GR, Jones CE, Trifilieff A (2003). IL‐17, produced by lymphocytes and neutrophils, is necessary for lipopolysaccharide‐induced airway neutrophilia: IL‐15 as a possible trigger. J Immunol 170: 2106–2112. [DOI] [PubMed] [Google Scholar]
  23. Fuentes JM, Fulton WB, Nino D, Talamini MA, Maio AD (2008). Atropine treatment modifies LPS‐induced inflammatory response and increases survival. Inflamm Res 57: 111–117. [DOI] [PubMed] [Google Scholar]
  24. Gosens R, Zagsma J, Meurs H, Halayko AJ (2006). Muscarinic receptor signaling in the pathophysiology of asthma and COPD. Respir Res 7: 93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Graham JR, Wright BS, Rezk PE, Gordon RK, Sciuto AM, Nambiar MP (2006). Butyrylcholinesterase in guinea pig lung lavage: a novel biomarker yo assess lung injury following inhalation exposure to nerve agent VX. Inhal Toxicol 18: 493–500. [DOI] [PubMed] [Google Scholar]
  26. Grommes J, Soehnlein O (2011). Contribution of neutrophils to acute lung injury. Mol Med 17: 293–307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Gu YJ, Wang YS, Chiang BY, Gao XD, Ye CX, Wildevuur CR (1991). Reduction of granulocyte‐mediated lung injury in canine cardiopulmonary bypass by anisodamine infusion. Thorac Cardiovasc Surg 39: 154–157. [DOI] [PubMed] [Google Scholar]
  28. Guo H‐Y, Lorenz RR, Vanhoutte PM (1992a). Anisodamine inhibits non‐selectively muscarinic receptors in isolated canine veins. Chin Med J (Engl) 105: 5–10. [PubMed] [Google Scholar]
  29. Guo H, Lorenz RR, Vanhoutte PM (1992b). Anisodamine antagonizes acetylcholine‐induced inhibition of adrenergic neurotransmission in the canine saphenous vein. Chin Med Sci J 7: 32–35. [PubMed] [Google Scholar]
  30. Guo HY, Lorenz RR, Vanhutte PM (1992c). Anisodamine inhibits acetylcholine‐induced endothelium‐dependent relaxation of canine femoral artery. Chin Med J (Engl) 105: 666–670. [PubMed] [Google Scholar]
  31. Guo HY, Lorenz RR, Vanhoutte PM (1993). Anisodamine at higher concentrations in inhibiting alpha‐adrenergic responses in isolated canine blood vessels. Chin Med J (Engl) 106: 452–457. [PubMed] [Google Scholar]
  32. Huang Q, Dai W, Jie Y, Yu G (2002). Protective effect of anisodamine on respiratory function after severe brain damage. Chin J Traumatol 5: 352–354. [PubMed] [Google Scholar]
  33. Hulse EJ, Davies JOJ, Simpson AJ, Sciuto AM, Eddleston M (2014). Respiratory complications of organophosphorous nerve agents and insecticide poisoning. Implications for respiratory and critical care. Am J Respir Crit Care Med 190: 1342–1354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Hwang F, Wang SM, Hu CC (1983). The disordering effect of hyoscyamine drugs on phospholipid membranes. Biochim Biophys Acta 736: 220–225. [DOI] [PubMed] [Google Scholar]
  35. Hwang GS, Hu S, Lin YH, Chen ST, Tang TK, Wang PS et al. (2013). Arecoline inhibits interlrukin‐2 secretion in Jurkat cells by decreasing the expression of alpha‐7‐nicotinic acetylcholine receptors and prostaglandin E2 . J Physiol Pharmacol (Krakow) 64: 535–543. [PubMed] [Google Scholar]
  36. Iyer R, Iken B, Leon A (2015). Development in alternative treatments for organophosphate poisoning. Toxicol Lett 233: 200–206. [DOI] [PubMed] [Google Scholar]
  37. Kaur S, Singh S, Chahal KS, Prakash A (2014). Potential pharmacological strategies for the improved treatment of organophosphate‐induced neurotoxicity. Can J Physiol Pharmacol 92: 893–911. [DOI] [PubMed] [Google Scholar]
  38. Kolahian S, Gosens R (2012). Cholinergic regulation of airway inflammation and remodeling. J Allerg Article ID 681258. doi:10.1155/2012/681258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Lavon O, Eisenkraft A, Blanca M, Raveh L, Ramaty E, Krivoy A et al. (2015). Is rivastigmine safe as pretreatment against nerve agents poisoning? A pharmacological, physiological and cognitive assessment in healthy young adult volunteers. Neurotoxicology 49: 36–44. [DOI] [PubMed] [Google Scholar]
  40. Li QB, Pan R, Wang GF, Tang SX (1999). Anisodamine as an effective drug to treat snake bites. J Nat Toxins 8: 327–330. [PubMed] [Google Scholar]
  41. Li Q, Lei H, Liu A, Yang Y, Su D, Liu X (2011). The antishock effect of anisodamine requires the upregulation of α7 nicotine acetylcholine receptors by IL‐10. Life Sci 89: 395–401. [DOI] [PubMed] [Google Scholar]
  42. Li CH, Zhang X, Ge XL, Huang X, Zhang AQ, Gu WQ (2014). Effects of combined anisodamine and neostigmine treatment on the inflammatory response and liver regeneration of obstructive jaundice in rats after hepatectomy. BioMed Res Int Article ID 362024. doi:10.1155/2014/362024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Liu C, Shen FM, Le YY, Kong Y, Liu X, Cai GJ et al. (2009). Antishock effect of anisodamine involves a novel pathway for activating alpha7 nicotinic acetylcholine receptor. Crit Care Med 37: 634–641. [DOI] [PubMed] [Google Scholar]
  44. Liu YH, Zhang J, Dai Z, Zhou MH, Li YH, Yin XL et al. (2013). Protection of anisodamine on the mitochondrial injury induced by oxidative stress in swine with cardiac arrest. Zhonghua Wei Zhong Bing Ji Jiu Yi Xue 25: 290–293. [DOI] [PubMed] [Google Scholar]
  45. Luo ZY, Xu I, Luo H (1987). Effect of anisodamine on glucocorticoid receptor of pulmonary alveolar macrophage in rabbits with oleic acid‐induced repiratory distress syndrome. Circ Shock 21: 349. [Google Scholar]
  46. Luo ZY, Tang Y, You JI, Luo H (1992). Protective effect of anisodamine on cultured bovine pulmonary endothelial cell injury induced by oxygen‐free radical. Arch Surg 127: 1204–1208. [DOI] [PubMed] [Google Scholar]
  47. Markel G, Krivoy A, Rotman E, Schein O, Shrot S, Brosh‐Nissimov T et al. (2008). Medical management of toxicological mass casualty events. Isr Med Assoc J 10: 761–766. [PubMed] [Google Scholar]
  48. Matthay MA, Ware LB, Zimmerman GA (2012). The acute respiratory distress syndrome. J Clin Invest 122: 2731–2740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Meduri GU, Headley AS, Golden E, Carson SJ, Umbarger RA, Kelso T et al. (1998). Effect of prolonged methylprednisolone therapy on unresolving acute respiratory distress syndrome. JAMA 280: 159–165. [DOI] [PubMed] [Google Scholar]
  50. Meduri GU, Tolley EA, Chrousos GP, Stentz F (2002). Prolonged methylprednisol; one treatment suppresses systemic inflammation in patients with unresolving acute respiratory distress syndrome. Evidence for inadequate endogenous glucocorticoid secretion and inflammation‐induced immune cell resistance to glucocorticoids. Am J Respir Crit Care Med 165: 983–991. [DOI] [PubMed] [Google Scholar]
  51. Meduri GU, Golden E, Freire AX, Taylor E, Zaman M, Carson SJ et al. (2007). Methylprednisolone infusion in early severe ARDS. Results of a randomized controlled trial. Chest 131: 954–963. [DOI] [PubMed] [Google Scholar]
  52. Menezes SLS, Bozza PT, Castro HC, Netto F, Laranjeira AP, Negri EM et al. (2005). Pulmonary and extrapulmonary acute lung injury: inflammatory and ultrastructural analyses. J Appl Physiol 98: 1777–1783. [DOI] [PubMed] [Google Scholar]
  53. Miyamoto M, Prause O, Sjöstrand M, Laan M, Lötvall J, Lindén A (2003). Endogenous IL‐17 as a mediator of neutrophil recruitment caused by endotoxin exposure in mouse airways. J Immunol 170: 4665–4672. [DOI] [PubMed] [Google Scholar]
  54. Moulton BC, Fryer AD (2011). Muscarinic receptor antagonists, from folklore to pharmacology; finding drugs that actually work in asthma and COPD. Br J Pharmacol 163: 44–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Nakagawa S, Kushiya K, Taneike I, Imanishi K, Uchiyama T, Yamamoto T (2005). Specific inhibitory action of anisodamine against a staphylococcal superantigenic toxin, toxic shock syndrome toxin 1 (TSST‐1), leading to down‐regulation of cytokine production and blocking of TSST‐1 toxicity in mice. Clin Diagn Lab Immunol 12: 399–408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Nambiar MP, Gordon RK, Rezk PE, Katos AM, Wajda NA, Moran TS et al. (2007). Medical countermeasures against respiratory toxicity and acute lung injury following inhalation exposure to chemical warfare agent VX. Toxicol Appl Pharmacol 219: 142–150. [DOI] [PubMed] [Google Scholar]
  57. Norby FL, Ren J (2002). Anisodamine inhibits cardiac contraction and intracellular Ca(2+) transients in isolated adult rat ventricular myocytes. Eur J Pharmacol 439: 21–25. [DOI] [PubMed] [Google Scholar]
  58. Pan SY, Han YF (2000). Learning deficits induced by four belladonna alkaloids are preferentially attenuated by tacrine. Acta Pharmacol Sin 21: 124–130. [PubMed] [Google Scholar]
  59. Pan SY, Han YF (2004). Comparison of the inhibitory efficacy of four belladonna drugs on gastrointestinal movement and cognitive function in food‐deprived mice. Int J Exp Clin Pharm 72: 177–183. [DOI] [PubMed] [Google Scholar]
  60. Pang Y‐H, Chen JW (2004). Anisodamine causes the changes of structure and function in the transmembrane domain of the Ca2+‐ATPase from sarcoplasmic reticulum. Biosci Biotechnol Biochem 68: 126–131. [DOI] [PubMed] [Google Scholar]
  61. Pawson AJ, Sharman JL, Benson HE, Faccenda E, Alexander SP, Buneman OP et al. , NC‐IUPHAR(2014). The IUPHAR/BPS guide to PHARMACOLOGY: an expert‐driven knowledge base of drug targets and their ligands. Nucleic Acids Res 42: D1098–D1106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Pavlov VA, Wang H, Czura CJ, Friedman SG, Tracey KJ (2003). The cholinergic anti‐inflammatory pathway: a missing link in neuroimmunomodulation. Mol Med 9: 125–134. [PMC free article] [PubMed] [Google Scholar]
  63. Perkins MW, Pierre Z, Rezk P, Song J, Marshall S, Oguntayo S et al. (2011a). Endotracheal aerosolization of atropine sulfate protects against soman‐induced acute respiratory toxicity in guinea pigs. Inhal Toxicol 23: 182–195. [DOI] [PubMed] [Google Scholar]
  64. Perkins MW, Pierre Z, Rezk P, Song J, Marshall S, Oguntayo S et al. (2011b). Protective effects of aerosolized scopolamine against soman‐induced acute respiratory toxicity in guinea pigs. Int J Toxicol 30: 639–649. [DOI] [PubMed] [Google Scholar]
  65. Perl M, Lomas‐Neira J, Venet F, Chung CS, Ayala A (2011). Pathogenesis of indirect (secondary) acute lung injury. Expert Rev Respir Med 5: 115–126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Poupko JM, Baskin SI, Moore E (2007). The pharmacological properties of anisodamine. J Appl Toxicol 27: 116–121. [DOI] [PubMed] [Google Scholar]
  67. Prause O, Bossios A, Silverpil E, Ivanov S, Bozinovski S, Vlahos R et al. (2009). IL‐17‐producing T lymphocytes in lung tissue and in the bronchoalveolar space after exposure to endotoxin from Escherichia coli in vivo‐effects of anti‐inflammatory pharmacotherapy. Pulm Pharmacol Ther 22: 199–207. [DOI] [PubMed] [Google Scholar]
  68. Qian J, Galitovskiy V, Chernyavsky AI, Marchenko S, Grando SA (2011). Plasticity of the murine spleen T‐cell cholinergic receptors and their role in in vitro differentiation of naïve CD4 T cells toward the Th1, Th2 and Th17 lineages. Genes Immun 12: 222–230. [DOI] [PubMed] [Google Scholar]
  69. Qian J, Zhang JM, Lin LL, Dong WZ, Cheng YQ, Su DF et al. (2015). A combination of neostigmine and anisodamine protects against ischemic stroke by activating α7nAChR. Int J Stroke 10: 737–744. [DOI] [PubMed] [Google Scholar]
  70. Razani‐Boroujerdi S, Behl M, Hahn FF, Pena‐Philippides JC, Hutt J, Sopori ML (2008). Role of muscarinic receptors in the regulation of immune and inflammatory responses. J Neuroimmunol 194: 83–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Rosas‐Ballina M, Tracey KJ (2009). Cholinergic control of inflammation. J Intern Med 265: 663–679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Rosman Y, Eisenkraft A, Milk N, Shiyovich A, Ophir N, Shrot S et al. (2014). Lessons learned from the Syrian sarin attack: evaluation of a clinical syndrome through social media. Ann Intern Med 160: 644–648. [DOI] [PubMed] [Google Scholar]
  73. Ruan QR, Zhang WJ, Hufnagl P, Kaun C, Binder BR, Wojta J (2001). Anisodamine counteracts lipopolysaccharide‐induced tissue factor and plasminogen activator inhibitor‐1 expression in human endothelial cells: contribution of the NF‐kappa B pathway. J Vasc Res 38: 13–19. [DOI] [PubMed] [Google Scholar]
  74. Ruan Q, Song J, Deng Z (2002). Anisodamine inhibits endotoxin‐induced tissue factor expression in human endothelial cells. J Huazhong Univ Sci Technolog Med Sci 22: 273–275 , 278. [DOI] [PubMed] [Google Scholar]
  75. Sawyer TW, Mikler J, Tenn C, Bjarnason S, Frew R (2012). Non‐cholinergic intervention of sarin nerve agent poisoning. Toxicology 294: 85–93. [DOI] [PubMed] [Google Scholar]
  76. Shytle RD, Mori T, Townsend K, Vendrame M, Sun N, Zeng J et al. (2004). Cholinergic modulation of microglial activation by alpha 7 nicotinic receptors. J Neurochem 89: 337–343. [DOI] [PubMed] [Google Scholar]
  77. Sidell FR, Newmark J, McDonough JH (2008). Chapter 5: Nerve Agents. In: Medical aspects of chemical warfare In: Tuorinsky SD. (ed). (Senior Editor)Textbook of military medicine. Office of the Surgeon General, US Army, Borden Institute, Walter Reed Army Medical Center: Washington DC, pp. 155–220. [Google Scholar]
  78. Song X, Pope C, Murthy R, Shaikh J, Lal B, Bressler JP (2004). Interactive effects of paraoxon and pyridostigmine on blood–brain barrier integrity and cholinergic toxicity. Toxicol Sci 78: 221–247. [DOI] [PubMed] [Google Scholar]
  79. Su JY, Hock CE, Lefer AM (1984). Beneficial effect of anisodamine in hemorrhagic shock. Naunyn Schmiedebergs Arch Pharmacol 325: 360–365. [DOI] [PubMed] [Google Scholar]
  80. Sun L, Zhang GF, Zhang X, Liu Q, Liu JG, Su DF et al. (2012). Combined administration of anisodamine and neostigmine produces anti‐shock effects: involvement of α7 nicotinic acetylcholine receptor. Acta Pharmacol Sin 33: 761–766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Testylier G, Lahrech H, Montigon O, Foquin A, Delacour C, Bernabé D et al. (2007). Cerebral edema induced in mice by a convulsive dose of soman. Evaluation through diffusion‐weighted magnetic resonance imaging and histology. Toxicol Appl Pharmacol 220: 125–137. [DOI] [PubMed] [Google Scholar]
  82. Varma DR, Yue TL (1986). Adrenoceptor blocking properties of atropine‐like agents anisodamine and anisodine on brain and cardiovascular tissues of rats. Br J Pharmacol 87: 587–594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Wang PY, Chen JW, Hwang F (1993). Anisodamine causes acyl chain interdigitation in phosphatidylglycerol. FEBS Lett 332: 193–196. [DOI] [PubMed] [Google Scholar]
  84. Wang H, Lu Y, Chen HZ (2003a). Differentiating effects of anisodamine on cognitive amelioration and peripheral muscarinic side effects induced by pilocarpine in mice. Neurosci Lett 344: 173–176. [DOI] [PubMed] [Google Scholar]
  85. Wang H, Yu M, Ochani M, Amella CA, Tanovic M, Susarla S et al. (2003b). Nicotinic acetylcholine receptor alpha7 subunit is an essential regulator of inflammation. Nature 421: 384–388. [DOI] [PubMed] [Google Scholar]
  86. Wang W, Chen QF, Ruan HL, Chen K, Chen B, Wen JM (2014a). Can anisodamine be a potential substitute for high‐dose atropine in cases of organophosphate poisoning? Hum Exp Toxicol 33: 1186–1190. [DOI] [PubMed] [Google Scholar]
  87. Wang W, Chen QF, Li QB, Wu YB, Chen K, Chen B et al. (2014b). Efficiency of anisodamine for organophosphorus‐poisoned patients when atropinization cannot be achieved with high doses of atropine. Environ Toxicol Pharmacol 37: 477–481. [DOI] [PubMed] [Google Scholar]
  88. Wen XM, Zhang YL, Liu XM, Guo SX, Wang H (2006). Immune responses in mice to arecoline mediated by lymphocyte muscarinic acetylcholine receptor. Cell Biol Int 30: 1048–1053. [DOI] [PubMed] [Google Scholar]
  89. Wessler I, Kirkpatrick CJ (2008). Acetylcholine beyond neurons: the non‐neuronal cholinergic system in humans. Br J Pharmacol 154: 1558–1571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Williams AE, Chambers RC (2014). The mercurial nature of neutrophils: still an enigma in ARDS? Am J Physiol Lung Cell Mol Physiol 306: L217–L230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Wu SH, Liao PY, Dong L, Chen ZQ (2008). Signal pathway involved in inhibition by lipoxin A(4) of production of interleukins induced in endothelial cells by lipopolysaccharide. Inflamm Res 57: 430–437. [DOI] [PubMed] [Google Scholar]
  92. Wu H, Li L, Su X (2014). Vagus nerve through α7 nAChR modulates lung infection and inflammation: models, cells, and signals. BioMed Res Int Article ID 283525. doi:10.1155/2014/283525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Xiao DM, Wang XL, Chen HC (1988). Inhibitory effects of anisodamine on endotoxin‐induced prostaglandin production by cells in culture. Chin Med J (Engl) 101: 871–875. [PubMed] [Google Scholar]
  94. Xing K, Fu X, Jiang L, Wang Y, Li W, Gu X et al. (2015). Cardioprotective effects of anisodamine against myocardial ischemia injury and its influence on cardiomyocyte apoptosis. Cell Biochem Biophys. doi:10.1007/s12013-015-0642-4. [DOI] [PubMed] [Google Scholar]
  95. Xiu R‐J, Hammerschmidt DE, Coppo PA, Jacob HS (1982). Anisodamine inhibits thromboxane synthesis, granulocyte aggregation and platelet aggregation. JAMA 247: 1458–1460. [DOI] [PubMed] [Google Scholar]
  96. Xu ZP, Wang H, Hou LN, Xia Z, Zhu L, Chen HZ et al. (2011). Modulatory effect of anisodamine on airway hyper‐reactivity and eosinophilic inflammation in a murine model of allergic asthma. Int Immunopharmacol 11: 260–265. [DOI] [PubMed] [Google Scholar]
  97. Xu ZP, Yang K, Xu GN, Zhu L, Hou LN, Zhang WH et al. (2012). Role of M3 mAChR in in vivo and in vitro models of LPS‐induced inflammatory response. Int Immunopharmacol 14: 320–327. [DOI] [PubMed] [Google Scholar]
  98. Yang P, Zhang BH, Hong N (1991). Anti‐arrhythmia and nervous system effects of anisodamine. Zhongguo Yao Li Xue Bao 12: 173–176. [PubMed] [Google Scholar]
  99. Yang XO, Chang SH, Park H, Nurieva R, Shah B, Acero L et al. (2008). Regulation of inflammatory responses by IL‐17F. J Exp Med 205: 1063–1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Yao T, Xiao YF, Sun XY, Tong HH, Yuan M, Xu ZY et al. (1984). Effects of anisodamine on cardiovascular activities in endotoxin‐shocked dogs. Chin Med J (Engl) 97: 871–876. [PubMed] [Google Scholar]
  101. You QH, Zhang D, Niu CC, Zhu ZM, Wang N, Yue Y et al. (2014). Expression of IL‐17 A and IL‐17F in lipopolysaccharide‐induced acute lung injury and the counteraction of anisodamine or methylprednisolone. Cytokine 66: 78–86. [DOI] [PubMed] [Google Scholar]
  102. Yu XY, Luo ZY, You JL, Luo H (1985). The influence of anisodamine on the cells from bronchoalveolar lavage in experimental respiratory distress syndrome. Bull Hunan Med Coll 10: 13–18. [Google Scholar]
  103. Zang SP, Yao L, Gugino SF, Russell JA (1987). Quantitative analysis of anisodamine‐acetylcholine antagonism in canine tracheal smooth muscle. Chin Med J (Engl) 100: 871–873. [PubMed] [Google Scholar]
  104. Zhang HM, Ou ZL, Yamamoto T (2001). Anisodamine inhibits shiga toxin type 2‐mediated tumor necrosis factor‐alpha production in vitro and in vivo. Exp Biol Med (Maywood) 226: 597–604. [DOI] [PubMed] [Google Scholar]
  105. Zhang JT (2002). New drugs from medicinal plants. Therapie 57: 137–150. [PubMed] [Google Scholar]
  106. Zhang T, Zhang X, Shao Z, Ding R, Yang S, Ruan J et al. (2005). The prophylactic and therapeutic effects of cholinolytics on perfluoroisobutylene inhalation induced acute lung injury. J Occup Health 47: 277–285. [DOI] [PubMed] [Google Scholar]
  107. Zhang WW, Song MK, Cui YY, Wang H, Zhu L, Niu YY et al. (2008). Differential neuropsychopharmacological influences of naturally occurring tropane alkaloids anisodamine versus scopolamine. Neurosci Lett 443: 241–245. [DOI] [PubMed] [Google Scholar]
  108. Zhang WW, Xu ZP, Cui YY, Wang H, Song MK, Li J et al. (2009). Peripheral cholinoceptor antagonist anisodamine counteracts cholinergic adverse effects and facilitates cognitive amelioration of rivastigmine. J Neural Transm 116: 1643–1649. [DOI] [PubMed] [Google Scholar]
  109. Zhao CL, Ron S, Liu CG, He XP, Xie ZP (1993). Blocking effect of anisodamine on acetylcholine receptor channels. Zhongguo Yao Li Xue Bao 14: 190–192. [PubMed] [Google Scholar]
  110. Zhao T, Li DJ, Liu C, Su DF, Shen FM (2011). Beneficial effects of anisodamine in shock involved cholinergic anti‐inflammatory pathway. Front Pharmacol 2: 23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Zhou JX, Ke P, Huan G, Shao BZ, Liu C (2014). Combined treatment with anisodamine and neostigmine inhibits joint inflammation in collagen‐induced arthritis in mice. CNS Neurosci Ther 20: 186–187. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from British Journal of Pharmacology are provided here courtesy of The British Pharmacological Society

RESOURCES