Sie sind auf Seite 1von 9

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/328793529

DARK Classics in Chemical Neuroscience: Opium, a Friend or Foe

Article  in  ACS Chemical Neuroscience · November 2018


DOI: 10.1021/acschemneuro.8b00546

CITATIONS READS

0 242

7 authors, including:

Parvez Alam Mohammad Khursheed Siddiqi


Aarhus University Aligarh Muslim University
49 PUBLICATIONS   1,041 CITATIONS    48 PUBLICATIONS   536 CITATIONS   

SEE PROFILE SEE PROFILE

Aquib Ehtram Nabeela Majid


Indian Institute of Technology Delhi Aligarh Muslim University
5 PUBLICATIONS   11 CITATIONS    5 PUBLICATIONS   5 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Protein misfolding and aggregation View project

Drug-Protein Interaction View project

All content following this page was uploaded by Parvez Alam on 16 November 2018.

The user has requested enhancement of the downloaded file.


Review

Cite This: ACS Chem. Neurosci. XXXX, XXX, XXX−XXX pubs.acs.org/chemneuro

DARK Classics in Chemical Neuroscience: Opium, a Friend or Foe


Parvez Alam,†,∥ Subhomoi Borkokoty,† Mohammad Khursheed Siddiqi,‡ Aquib Ehtram,†
Nabeela Majid,‡ Moin Uddin,§ and Rizwan Hasan Khan*,‡

Kusuma School of Biological Sciences, Indian Institute of Technology, Hauz Khas, New Delhi 110016, India

Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India
§
Department of IlmulAdvia (Unani Pharmacology), Ajmal Khan Tibbiya College, Aligarh Muslim University, Aligarh 202002, India

ABSTRACT: Opium has found great use medicinally for its analgesic properties and
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

has been witnessed as one of the most popular medications used in psychiatry. Opium
derivatives have been shown as efficacious for relieving pain and the treatment of
epileptic seizures, but progressive research toward their use in the treatment of
neurodegenerative diseases remain elusive. To gain more insight into the other
Downloaded via AARHUS UNIV on November 15, 2018 at 11:54:10 (UTC).

properties of opium such as anti-inflammatory properties, herein we discuss basic


information regarding opium, opium content and mechanism of action, pharmacology of
opium derivatives, the role of opium in the prevention of neurodegeneration, and
adverse effects of opium derivatives on neuronal health.

KEYWORDS: Opium, neuroscience, neurodegeneration, medicine, mechanism

■ INTRODUCTION
Opium is the dried latex obtained from the opium poppy.
(1st century) and Galen (129−199) have referred to the
narcotic analgesic properties of opium.1 And it was Paracelcus
Opium latex contains morphine, codeine, thebaine, papaverine (1493−1541) a Swiss German alchemist who observed that
,and noscapine (Figure 1). Since prehistoric times, opiates have certain analgesic opium alkaloids are far more soluble in
been used for medicinal purposes including relieving pain in alcohol than in water which led to tinctura laudanum, allowing
cancer, spasms from tetanus, and pain associated with
menstruation and childbirth. The plant papaver has been for easy medicinal delivery2,3 and thus paving the way for
known for its medicinal benefits as have several other opium’s regimented use in medicine. In the following century,
benzylisoquinoline alkaloids (BIAs) with potent pharmaco- Thomas Sydenham (1624−1689) recommended opium
logical properties including the vasodilator papaverine, the against hysteria and mania.4 Then, in general, the 18th century
cough suppressant and potential anticancer drug noscapine, witnessed opium as one of the more popular medications used
and the antimicrobial agent sanguinarine. The collection of
in psychiatry.5−7 In 1991, Weber and Emrich published an
opium has been active since approximately 3400 BCE. The
oldest archeological occurrence of poppy species was extraordinary review of opiate treatment in psychiatric
documented for the wild poppy, Papaver setigerum. Many disorders.8
alkaloids are being derived from opium but the only important Previously, our group has reported the effect of small
opiates include morphine (10%) and codeine. Morphine was molecules in preventing amyloid formation in proteins and
identified first as the active pharmacological ingredient of their ability to protect neuronal cells against amyloid induced
opium (making up between 9% and 14% of opium) in the early
1800s and was then consumed by patients with malaria for cytotoxicity.9−15 In this Review, we discuss some basic
pain relief because it was cheap and easily available although information regarding opium, opium content and mechanism
the quinones specific for malaria treatment were present at that of action, pharmacology of opium derivatives, the role of
time but they were not affordable. Heroin (diacetylmorphine) opium in prevention of neurodegeneration, and adverse effects
was further synthesized from morphine modifications in the of opium derivatives on the neuronal health, and we attempt to
late 1800s in an attempt to develop a nonaddicting cough
widely disperse data into one easily accessible format for the
suppressant which was 5−10 times more potent as morphine
yet safer. Papaverine, an opium derivative, is occasionally used “Classics in Chemical Neuroscience” series.
as a vasodilator.
In the Odyssey, Homer refers to a curative substance, which Received: October 10, 2018
was administered to Helena as a remedy against grief and Accepted: November 7, 2018
grudge. Similarly, the classical medical writings of Dioscurides Published: November 7, 2018

© XXXX American Chemical Society A DOI: 10.1021/acschemneuro.8b00546


ACS Chem. Neurosci. XXXX, XXX, XXX−XXX
ACS Chemical Neuroscience Review

Figure 1. Structures of the most common opium alkaloids.

Figure 2. Experimental structures of the classical opioid receptors: KOP (PDB ID: 4DJH),30 MOP (PDB ID: 5C1M),31 and DOP (PDB ID:
4N6H)32 with NOP (PDB ID: 4EA3)33 and their major targets in the nervous system.

■ OPIUM CONTENT AND MECHANISM OF ACTION


Opium contains both nonalkaloid and alkaloid constituents.
pain neurotransmitters.18 Opioids reduce the intensity and
unpleasantness of pain. They produce their effects by activating
The nonalkaloid part is made up of water (∼5−20%), various specific G protein-coupled receptors in the brain, spinal cord,
sugars (∼20%), and several simple organic acids such as and peripheral nervous system.
fumaric acid, lactic acid, oxaloacetic acid, and meconic acid. Opioids have specific binding to distinct receptors. Based on
While the alkaloid content is approximately 10−20% with their differences in opioid potency, selective antagonism, and
more than 40 individual alkaloids including major alkaloids stereospecificity of opiate actions, opioid receptors (OPs) can
such as morphine, codeine, thebaine, etc. and minor alkaloids be classified into three major classes based on their affinity for
such as aporphines, rhoeadines, tetrahydroisoquinolines, etc.16 various opioid ligands and in their distribution in the nervous
Opioids can be classified based on their source, chemical system: δ-opioid (DOP), κ-opioid (KOP), and μ-opioid
structure, and function.17,18 Opioids can act as agonists, (MOP) (Figure 2). These are termed as classical opioid
antagonists, agonists/antagonists, or partial agonists. Agonist receptors.18,19 However, there is a fourth type of OP termed as
activity at opioid receptors acts to open potassium channels nonclassical, i.e. nociceptin/orphanin FQ (N/OFQ) peptide
and prevent the opening of voltage-gated calcium channels. receptor (NOP). NOP shares significant sequence homology
This reduces neuronal excitability and inhibits the release of with the classical opioid receptors.20 The ε-, ι-, λ-, and ζ-
B DOI: 10.1021/acschemneuro.8b00546
ACS Chem. Neurosci. XXXX, XXX, XXX−XXX
ACS Chemical Neuroscience Review

receptors are few of the novel but poorly characterized opioid


receptors. Initially considered to be an opioid receptor, the σ-
■ MEDICINAL CHEMISTRY AND PHARMACOLOGY
OF OPIUM
receptor is no longer classified with the opioid receptors As we have seen in Table 1, opioids can be classified as agonist,
because it displays neither the stereoselectivity characteristic of antagonist, partial agonist, or antagonist and mixed action.
opioid receptors nor antagonism by opioid antagonists.21
OPs are coupled to G proteins (G1 and G0), and they Table 1. Classification of Opioids
interact with these G proteins to inhibit adenylate cyclase, an
enzyme that promotes the formation of the intracellular source natural morphine, codeine
messenger cyclic adenosine 3′,5′-monophosphate (cyclic semisynthetic tramadol, heroin, oxycodone,
oxymorphone, and buprenorphine
AMP). G proteins also may mediate the inhibitory effects of synthetic entanyl, pethidine, and
opioids on voltage-gated calcium channels and their activating dextropropoxyphene
effects on inwardly rectifying potassium channels. Inhibition of chemical phenanthrenes morphine, codeine, heroin,
calcium channels may lead to a decrease in the release of structure hydromorphone, and oxycodone
neurotransmitters such as substance P, which mediates pain benzomorphans pentazocine and phenazocine
diphenylpropylamines propoxyphene, methadone, levo-Α-
signals.22 acetylmethadol, and loperamide
DOPs (agonist delta-alanine-delta-leucine-enkephalin) are phenylpiperidines meperidine
distributed throughout the central nervous system (CNS). anilidopiperidines fentanyl, alfentanil, and sufentanil
DOP density varies in different brain regions: the highest oripavine derivatives etorphine, dihydroetorphine, and
densities can be found in the olfactory bulb, neocortex, caudate buprenorphine
putamen, and nucleus accumbens, and to a lesser degree the morphinan derivatives levorphanol and butorphanol
thalamus, hypothalamus, and brain stem.20 This receptor is synthetic analogue of tramadol
codeine
involved in the antinociceptive/analgesic actions of some
function pure agonists morphine and codeine
opioids. Activation of DOP results in decreased presynaptic
partial agonists buprenorphine
Ca2+ influx, which inhibits release of neurotransmitters, and an mixed action pentazocine, nalbupine, and
increase in postsynaptic K+ efflux, which hyperpolarizes the butorphanol
neuron.23 antagonists naxolone
KOPs (agonist ketocyclazocine) are widely expressed
throughout the brain, spinal cord, and peripheral tissues. Their activity differs owing to their action on different opioid
They are responsible for spinal analgesia, sedation, dyspnea, receptors as well as genetic differences in opioid receptor
dependence, dysphoria, and respiratory depression. Activation sensitivity.24
of KOPs primarily leads to an inhibition of adenylyl cyclase Morphine, a classic example of a natural opioid, has agonistic
through the Gα subunit and induces increased potassium actions at the μ-, κ-, and δ-receptors.16 Being on the World
channel conductance and decreased calcium conductance via Health Organization (WHO) essential drugs list, it is widely
the Gβγ subunit. Modulation of these ion channels results in used as an analgesic commonly administered by different
decreased action potential generation and neurotransmitter routes such as intramuscular, intravenous, or oral. Major
release.24,25 features of morphine and morphine-like agonists in the CNS
MOP receptors are located throughout the central nervous include analgesia, drowsiness, euphoria, respiratory depression,
system including the cerebral cortex, amygdala (of the limbic nausea and vomiting, depressed cough reflex, and hypo-
system), and putamen of the basal ganglia (highest density).26 thermia.16,34 Morphine (285.34 g mol−1) has an oral
MORs modulate intracellular effectors through inhibitory Go/ bioavailability of 30%−75%, plasma elimination half-life of
Gi proteins as opioid agonists bind. In turn, receptor signaling 2−3.5 h, and octanol−water partition coefficient of 0.7.
is readily terminated by several cellular regulatory processes Morphine is chiefly metabolized through conjugation reactions
such as phosphorylation, desensitization, endocytosis, and with UGT2B7 (UDP-glucuronosyltransferase-2B7) enzyme,
downregulation.27 Antagonists like naloxone and naltrexone and hence, any genetic polymorphism of UGT2B7 may affect
block MOP, whereas agonists like methadone and buprenor- the action of morphine.35 The contributor of analgesic activity
phine partially activate the MOP.27 Major side effects of morphine is an active metabolite of the UGT2B7 pathway,
morphine-6-glucuronide (M-6-G). M-6-G is responsible for
associated with MOP agonists include respiratory depression,
sedation and nausea in patients with impaired renal excretory
while with MOP antagonist side effects such as peristalsis and
function (as it is excreted in the urine).34,36 A more water-
constipation are common. MOP opioids also have effects on soluble morphine metabolite (octanol−water partition coef-
the cardiovascular system, thermoregulation, hormone secre- ficient of 1.28), hydromorphone, is eight times more active
tion, and immune function.28 than morphine, and it does not have an active renally
The NOP receptor sequence is approximately 50−60% eliminated metabolite.16,34
identical to the classic opioid receptors.29 N/OFQ and its Naloxone (327.38 g mol−1) is a competitive μ-receptor
receptor are widely expressed throughout both the central and antagonist to opioids in the central nervous system. Naloxone
the peripheral nervous system.20 The N/OFQ (O, orphanin; F, also has been shown as an antagonist at other opioid receptors
phenylalanine; and Q, glutamine) peptide activates the NOP κ and δ but with a higher affinity for the μ receptor.24 Used as
receptor and triggers intracellular signaling events, including an antidote for opioid overdose, it has parenteral, intranasal,
inhibition of adenylyl cyclase and activation of various kinases pulmonary, or orotracheal intubation route of administration.37
like protein kinase C (PKC) and p38 mitogen-activated Naloxone is a potent inhibitor of the UGT2B7 pathway and
protein kinase and modulation of calcium and potassium thereby reduces the effect of morphine by altering M-6-G
channel conductance.29 production; however, naloxone has no effect if opioids are not
C DOI: 10.1021/acschemneuro.8b00546
ACS Chem. Neurosci. XXXX, XXX, XXX−XXX
ACS Chemical Neuroscience Review

present in a person.24 In naloxone postoperative patients, roles in attenuating neurodegeneration. Nootropic roles of
various adverse effects include cardiac disorders, gastro- opium derivatives are presented in Table 2 and discussed
intestinal disorders, nervous system disorders, psychiatric thereafter in detail.
disorders, respiratory, thoracic and mediastinal disorders, skin
and subcutaneous tissue disorders, and vascular disorders.37 Table 2. Opium Derivatives and Their Nootropic Effect
Naloxone is metabolized in the liver and primarily undergoes
glucuronidation to form naloxone-3-glucuronide and is opium nootropic effect
derivatives
excreted through urine (24−37% excreted as metabolites
within first 6 h).38 Naloxone has low oral bioavailability (∼2%) DM
analogues
DM and DX have anticonvulsant effects that suppress seizures
induced by electroshock51−53
due to extensive first-pass metabolism.39,40 DM prevents seizures, mortality, and hippocampal cell loss in a
The mixed agonist−antagonist and partial agonist bupre- dose-dependent manner54
norphine (467.64 g mol−1) is a semisynthetic opioid, has a DM minimizes the toxicity of the glutamate on the neuronal
poor bioavailability with extensive first pass effect by the liver, cells54
but has excellent sublingual bioavailability due to high lipid DX attenuates both morphological and chemical evidence of
glutamate neurotoxicity in murine neocortical cell cultures54
solubility.16,39 This potent analgesic is a high-affinity partial μ-
3-HM neuroprotective against endotoxin-induced DA neurotoxicity55
receptor agonist and high-affinity δ- and κ-receptor antagonist
nootropic to DA neurons in primary mixed mesencephalic
and has moderate affinity at NOP receptors.41 As an agonist, neuro-glial cultures56
buprenorphine is 30 times more potent than morphine. It also causes the release of certain nootropic factors (i.e., EGF,
has low oral bioavailability due to extensive first-pass GDNF, TGF-β1, TGF-α1, and ADNF) from astroglia,
metabolism and hence is administered through sublingual exerting nootropic effect on DA neurons55
mode.18 The primary mode of elimination of buprenorphine is most potent in restoring DA neuronal loss and DA depletion as
well as in attenuating behavioral damage55
through feces, with approximately 10−30% excreted in urine.42
increases the levels of nootropic factors and decreases the
Common buprenorphine adverse effects may include sedation, production of reactive oxygen species in LPS and MPTP PD
nausea and/or vomiting, dizziness, headache, and respiratory models55
depression.24 Buprenorphine is also used as one of the naloxone attenuate microglial activation in a dose-dependent manner57
medications to manage withdrawal from opioid drugs.43 attenuates cytokines that block the astrocyte activation,
Butorphanol (327.46 g mol−1) is an example of mixed decreasing the brain vulnerability to epilepsy58
agonist-antagonist opioid analgesic. It has an affinity for μ-, δ-, inhibits microglia driven neural inflammation59
and κ-opioid receptor subtype. Butorphanol exhibits pure
agonistic activity at the κ-receptor and antagonistic activity at DM Analogues. Opium derivatives have also been found
the δ − receptor, while at μ-receptor it acts as a partial to be involved in the treatment of epileptic seizures known as
agonist.44 As an agonist, subcutaneous butorphanol is 3 to 5 anticonvulsants. Dextromethorphan (DM; 3-methoxy-17-
times as potent as morphine and as an antagonist 15 to 25 methylmorphinan) and its metabolite dextrorphan (DX)
times as potent as mixed agonist-antagonist opioid analgesic, have been reported to have anticonvulsant effects that suppress
pentazocine in human.45 Butorphanol is extensively metabo- seizures induced by electroshock.51−53 Most studies confirm
lized in humans, mainly to hydroxybutorphanol. Butorphanol that DX is several times more potent than DM as an
is excreted through the urinary (about 70%) and biliary (about anticonvulsant.51 This may be due to the in vivo conversion of
11−14%) mode of elimination. Adverse effects include DM to DX significantly contributing toward the anticonvulsant
sedation, nausea, respiratory depression, and psychotomimetic activity of the parent molecule. Both molecules show an affinity
reactions.45 toward σ-receptors, the binding that is responsible for the
Tramadol (263.381 g mol−1), a synthetic 4-phenyl- anticonvulsant properties. DM has 2−5-fold higher affinity
piperidine analogue of codeine, acts on the μ-opioid receptor than DX, whereas the anticonvulsant property of DX is higher
as a weak agonist.46,47 However, its metabolite M1 has 300 than that of DM.60 Thus, the mechanism underlying the
times more affinity for the μ-opioid receptor as compared to anticonvulsant effect remains undetermined. Both anticonvul-
the parent compound of tramadol. The other mechanisms by sive and proconvulsive actions have been reported for the σ-
which tramadol acts on the central nervous system, including receptor binding that is responsible for the neuroprotective
inhibition of reuptake of serotonin and norepinephrine, are behavior.51
contributed by the enantiomers of tramadol, (+)-tramadol and Earlier studies have suggested that DM prevents seizures,
(−)-tramadol, respectively.47,48 Tramadol and its metabolites mortality, and hippocampal cell loss in a dose-dependent
are mainly excreted via the kidneys.49 Tramadol is used manner. This could be explained by the anticonvulsant
primarily as an analgesic but is also seen as a useful agent in the properties of the molecule that minimizes the toxicity of the
management of heroin withdrawal.43 The extended-release glutamate on the neuronal cells by inhibiting NMDA
(ER) formulation of tramadol is more effective than clonidine receptors.54 DM manages to inhibit NMDA receptor by
and comparable to buprenorphine in reducing opioid with- acquiring a noncompetitive antagonist site of the receptor. On
drawal symptoms.50


the contrary, DX is a more potent ligand toward the
noncompetitive antagonist site of the NMDA receptor and is
NOOTROPISM OF OPIUM DERIVATIVES constant with the anticonvulsant effects in vivo.54,61
Nootropic is a term used for the exploitation of medications or The dextrorotatory morphinan opioid, dextrorphan, which
wholesome supplements that positively affect brain function. has recently been reported to block the excitation of cortical
Various pharmaceutical compounds are available in the market, neurons by N-methyl-D-aspartate, was found at 10−100 μM
which have been exploited for their neuroprotective properties concentrations to attenuate both morphological and chemical
by altering specific neurotransmitters or opioid receptor evidence of glutamate neurotoxicity in murine neocortical cell
binding. Several opium derivatives have been found to play cultures.54
D DOI: 10.1021/acschemneuro.8b00546
ACS Chem. Neurosci. XXXX, XXX, XXX−XXX
ACS Chemical Neuroscience Review

3-HM. Progressive research for the development of effective Endotoxin driven inflammation leads to the activation of
therapeutics against neurodegeneration is limited. A structural microglia, which in turn produces proinflammatory cytokines
analogue of DM, 3-HM, which lacks methyl groups at O and N and neurotoxic factors, such as TNF-α, NO, IL-10, superoxide,
sites, showed prominent results in the treatment of Parkinson’s and free radicals, which subsequently mediates dopaminergic
disease (PD) and showed neuroprotective properties among neurodegeneration.68 Naloxone has been shown to be a
the DM analogues available.55 Studies showed that 3-HM was neuroprotective agent through the inhibition of microglia
more potent in restoring neuroprotection against endotoxin- driven neural-inflammation.59 The underlying mechanism lies
induced neurotoxicity than DM, its parent molecule.56 The with the inhibition of cytokines and neurotoxic factors from
nootropic effect of 3-HM was specifically glial dependent, and activated microglial cells.69


3-HM failed to restore any protective impact in neuron-
enriched cultures. 3-HM was neuroprotective against endotox- DARK SIDE OF OPIUM DERIVATIVES IN
in-induced dopamine (DA) neurotoxicity and also to DA NEURODEGENERATION
neurons in primary mixed mesencephalic neuro-glial cul-
tures.55,56 There are several reports claiming the induction of apoptosis
Furthermore, 3-HM caused the release of certain nootropic by the majority of opium derivatives. High doses of morphine
factors (i.e., EGF, GDNF, TGF-β1, TGF-α1, and ADNF) from during chronic treatment induce apoptosis in both neurons
astroglia, exerting neuroprotective effect of 3-HM on DA and microglial cells.70 The cytotoxicity of morphine has been
neurons. Glial cells, especially astroglia, protect stressed DA found to be dose-dependent where a single dose of morphine
neurons through the production of various nootropic factors did not induce apoptosis but the administration of large
that counter oxidative stress in substantia nigra pars in PD.55 morphine doses induced apoptosis in neuronal cells.70,71 The
One potential therapeutic intervention will be to stimulate morphological studies showed that chronic morphine treat-
astroglia to produce these nootropic factors to rescue damaged ment leads to substantial injuries in cerebral cortex and
neurons.55 The anti-inflammatory mechanism of 3-HM was hippocampus. This eventually contributed to the reduction in
attributed to its inhibition of endotoxin-induced production of dendritic complexity and decreased dendritic growth in the
an array of proinflammatory and neurotoxic factors. Thus, 3- cerebral cortex and hippocampus72
HM provides neuroprotection by acting on two different cell Based on animal model studies, the most symptoms
targets: a nootropic effect mediated by astroglia and an anti- observed in DM abusers are caused by DX, a major metabolite,
which binds to the same central nervous system receptors as a
inflammatory effect mediated by inhibition of microglial
major metabolite of DM, phencyclidine (PCP). Hyper-
activation.55
locomotor activity is observed in the mice offspring when
Several investigations for the neuroprotective property of
exposed to DM during prenatal stages. Marked behavioral
analogues of dextromethorphan (DM) in endotoxin-lip-
depression is demonstrated when these offspring received
opolysaccharide (LPS) and 1-methyl-4-phenyl-1,2,3,6-tetrahy-
additional DM.51,62
dropyridine (MPTP) models to classify neuroprotective drugs
Pathological damage in the posterior cingulate cortex and
for PD have been done.55 In vivo studies on the two models
retrosplenial cortex of the rat brain has been reported with a
showed that daily doses of DM metabolite protected DA
single treatment with noncompetitive NMDA receptor
neurons in substantia nigra pars compacta and restored DA
antagonists such as MK-801, PCP, or ketamine. In contrast,
levels in striatum. 3-HM, was found to be the most effective in
oral administration of high doses of DM does not produce
restoring DA neuronal loss and DA depletion as well as in
neuropathological changes as reported for other NMDA
attenuating behavioral damage. By increasing the gene
receptor antagonists, suggesting that there are route-specific
expression of various nootropic factors, and decreasing the
effects on the disposition of DM and its metabolites in rat
production of ROS (reactive oxygen species), 3-HM showed
brain73


the neuroprotective properties in LPS and MPTP PD
models.55,62 Due to its high efficacy and low toxicity, 3-HM
could be a novel therapy for PD. CONCLUSION
Naloxone. The role of inflammation has been long Opium derivatives act as potential agents toward neuro-
established in the groups of epileptic neurological disorders.63 protection. In this Review, the mechanism of their action has
Primary symptoms commonly includes episodes of epileptic been evaluated that supports the idea that opium derivatives
seizures. Rapid astrocyte and microglial activation precedes the upon binding to their respective receptors cause the decreased
seizures and hippocampal neurodegeneration which plays a influx of calcium which can be exploited for the treatment of
significant role in epileptogenesis.64,65 The precise mechanism neurodegenerative diseases such as Alzheimer’s disease and
of microglial activation still remains elusive. Cytokine Parkinson’s disease. As we know there occur rapid and
production is a common phenomenon by glia when the sustained increase in cytosolic calcium level because of amyloid
seizures are associated with neuronal injury.66 Status oligomer formation during neurodegenerative diseases. The
epilepticus (SE) induced cytokine production in postnatal increased calcium level also triggers the rapid transformation of
day 15 (PN 15) was studied, where IL-1β and S-100β levels protofibrils into mature amyloid fibrils. Despite their negative
were found to be strongly alleviated.58,67 Naloxone, a potential effects on overdosage that are comparatively less than their
opioid receptor antagonist, has been investigated to attenuate beneficial effects, they can be included by pharmacists in the
microglial activation in a dose-dependent manner.57 The formulation of antiaggregating drugs for the treatment of
studies suggested PN 15 SE induced glial derived IL-1β and S- various neurodegenerative diseases. There is an urgent need to
100β could be minimized when given in optimal doses.58 design drug molecules based on opium contents, and their
Naloxone driven attenuation of cytokines blocks the astrocyte neuroprotective potential must be explored in animal models
activation, thereby decreasing the brain vulnerability to in order to have improved therapy for neurodegenerative
epilepsy.58 diseases in future.
E DOI: 10.1021/acschemneuro.8b00546
ACS Chem. Neurosci. XXXX, XXX, XXX−XXX
ACS Chemical Neuroscience Review

■ ASSOCIATED CONTENT
Special Issue Paper
(2018) Stabilizing proteins to prevent conformational changes
required for amyloid fibril formation. J. Cell. Biochem.,
DOI: 10.1002/jcb.27576.
Published as part of the special issue “DARK Classics in (13) Siddiqi, M. K., Alam, P., Chaturvedi, S. K., Khan, M. V., Nusrat,
Chemical Neuroscience”.


S., Malik, S., and Khan, R. H. (2018) Capreomycin inhibits the
initiation of amyloid fibrillation and suppresses amyloid induced cell
AUTHOR INFORMATION toxicity. Biochim. Biophys. Acta, Proteins Proteomics 1866 (4), 549−
Corresponding Author 557.
*E-mail: rizwanhkhan1@gmail.com. Phone: +91-571-2720388. (14) Alam, P., Siddiqi, M. K., Chaturvedi, S. K., Zaman, M., and
Khan, R. H. (2017) Vitamin B12 offers neuronal cell protection by
Fax: + 91-571-2721776.
inhibiting Aβ-42 amyloid fibrillation. Int. J. Biol. Macromol. 99, 477−
ORCID 482.
Rizwan Hasan Khan: 0000-0002-9965-8982 (15) Chaturvedi, S. K., Alam, P., Khan, J. M., Siddiqui, M. K.,
Present Address Kalaiarasan, P., Subbarao, N., Ahmad, Z., and Khan, R. H. (2015)
∥ Biophysical insight into the anti-amyloidogenic behavior of taurine.
P.A.: Department of Biomedicine, Aarhus University, Aarhus
Int. J. Biol. Macromol. 80, 375−384.
C-8000, Denmark. (16) Schiff, P. L. (2002) Opium and its alkaloids. Am. J. Pharm.
Funding Educ. 66 (2), 186.
For providing financial assistance M.K.S. and S.B. are thankful (17) Akhgari, M., Etemadi-Aleagha, A., and Jokar, F. (2016) Street
to Department of Biotechnology (DBT), New Delhi, India. level heroin, an overview on its components and adulterants. In
P.A., A.E., and N.M. are thankful to Council of Scientific and Neuropathology of Drug Addictions and Substance Misuse 1, 867−877.
Industrial Research (CSIR), New Delhi, India. R.H.K. is (18) Lee, M. C., and Abrahams, M. (2012) Pain and analgesics. In
thankful to CSIR and UGC for project referenced as Clinical Pharmacology, 11th ed., pp 278−294.
(19) Reisine, T., and Bell, G. I. (1993) Molecular biology of opioid
37(1676)/17/EMR − II and F. 19-219/2018, respectively. receptors. Trends Neurosci. 16 (12), 506−10.
Notes (20) McDonald, J., and Lambert, D. G. (2013) Opioid mechanisms
The authors declare no competing financial interest. and opioid drugs. Anaesthesia & Intensive Care Medicine 14 (11), 505−

■ ACKNOWLEDGMENTS
Facilities provided by Kusuma School of Biological Sciences,
509.
(21) Corbett, A. D., Henderson, G., McKnight, A. T., and Paterson,
S. J. (2006) 75 years of opioid research: the exciting but vain quest for
the Holy Grail. Br. J. Pharmacol. 147 (Suppl 1), S153−62.
Indian Institute of Technology, HauzKhas, New Delhi, and (22) Knapp, C. M. (2002) Opiates. In Encyclopedia of the Human
Interdisciplinary Biotechnology Unit, Aligarh Muslim Uni- Brain, pp 729−739.
versity, Aligarh are gratefully acknowledged.


(23) Peppin, J. F., and Raffa, R. B. (2015) Delta opioid agonists: a
concise update on potential therapeutic applications. J. Clin. Pharm.
REFERENCES Ther. 40 (2), 155−66.
(1) Salerian, A. J. (2016) Endorphin agonists for psychiatric (24) Trescot, A. M., Datta, S., Lee, M., and Hansen, H. (2008)
disorders. Phys. Med. Rehabil. Res. 1 (3), 58−14. Opioid pharmacology. Pain Physician 11 (2 Suppl), S133−53.
(2) Salerian, A. J. (2015) Endorphin agonists for severe depression. (25) Wang, Y. H., Sun, J. F., Tao, Y. M., Chi, Z. Q., and Liu, J. G.
Pharm. Pharmacol. Int. J. 2 (2), 49−56. (2010) The role of kappa-opioid receptor activation in mediating
(3) Heymans, C. (1967) Pharmacology in old and modern antinociception and addiction. Acta Pharmacol. Sin. 31 (9), 1065−70.
medicine. Annu. Rev. Pharmacol. 7 (1), 1−15. (26) Liao, D., Lin, H., Law, P. Y., and Loh, H. H. (2005) Mu-opioid
(4) Park, R. H. R., and Park, M. P. (1990) Saint Vitus’ dance: vital receptors modulate the stability of dendritic spines. Proc. Natl. Acad.
misconceptions by Sydenham and Bruegel. J. R. Soc. Med. 83 (8), Sci. U. S. A. 102 (5), 1725−30.
512−515. (27) Contet, C., Kieffer, B. L., and Befort, K. (2004) Mu opioid
(5) Lader, M. (1980) Benzodiazepines-The opium of the masses? In receptor: a gateway to drug addiction. Curr. Opin. Neurobiol. 14 (3),
Commentaries in the Neurosciences, pp 609−615, Elsevier. 370−8.
(6) Ng, S. V. (2016) Opium Use in 19th-Century Britain: The Roots (28) McDonald, J., and Lambert, D. G. (2005) Opioid receptors.
of Moralism in Shaping Drug Legislation. American Journal of Continuing Education in Anaesthesia Critical Care & Pain 5 (1), 22−25.
Psychiatry Residents’ Journal 11 (6), 14−14. (29) Donica, C. L., Awwad, H. O., Thakker, D. R., and Standifer, K.
(7) Weber, M. M., and Emrich, H. M. (1988) Current and historical M. (2013) Cellular mechanisms of nociceptin/orphanin FQ (N/
concepts of opiate treatment in psychiatric disorders. International OFQ) peptide (NOP) receptor regulation and heterologous
clinical psychopharmacology 3 (3), 255−266. regulation by N/OFQ. Mol. Pharmacol. 83 (5), 907−18.
(8) Emrich, H. M., and Schmauss, C. (1991) Psychiatric aspects of (30) Wu, H., Wacker, D., Mileni, M., Katritch, V., Han, G. W.,
opioid research. In Neurobiology of opioids, pp 363−367, Springer. Vardy, E., Liu, W., Thompson, A. A., Huang, X. P., Carroll, F. I.,
(9) Alam, P., Chaturvedi, S. K., Siddiqi, M. K., Rajpoot, R. K., Ajmal, Mascarella, S. W., Westkaemper, R. B., Mosier, P. D., Roth, B. L.,
M. R., Zaman, M., and Khan, R. H. (2016) Vitamin k3 inhibits Cherezov, V., and Stevens, R. C. (2012) Structure of the human
protein aggregation: implication in the treatment of amyloid diseases. kappa-opioid receptor in complex with JDTic. Nature 485 (7398),
Sci. Rep. 6, 26759. 327−32.
(10) Alam, P., Beg, A. Z., Siddiqi, M. K., Chaturvedi, S. K., Rajpoot, (31) Huang, W., Manglik, A., Venkatakrishnan, A. J., Laeremans, T.,
R. K., Ajmal, M. R., Zaman, M., Abdelhameed, A. S., and Khan, R. H. Feinberg, E. N., Sanborn, A. L., Kato, H. E., Livingston, K. E.,
(2017) Ascorbic acid inhibits human insulin aggregation and protects Thorsen, T. S., Kling, R. C., Granier, S., Gmeiner, P., Husbands, S. M.,
against amyloid induced cytotoxicity. Arch. Biochem. Biophys. 621, Traynor, J. R., Weis, W. I., Steyaert, J., Dror, R. O., and Kobilka, B. K.
54−62. (2015) Structural insights into micro-opioid receptor activation.
(11) Siddiqi, M. K., Alam, P., Chaturvedi, S. K., and Khan, R. H. Nature 524 (7565), 315−21.
(2016) Anti-amyloidogenic behavior and interaction of Diallylsulfide (32) Fenalti, G., Giguere, P. M., Katritch, V., Huang, X. P.,
with Human Serum Albumin. Int. J. Biol. Macromol. 92, 1220−1228. Thompson, A. A., Cherezov, V., Roth, B. L., and Stevens, R. C. (2014)
(12) Siddiqi, M. K., Alam, P., Malik, S., Majid, N., Chaturvedi, S. K., Molecular control of delta-opioid receptor signalling. Nature 506
Rajan, S., Ajmal, M. R., Khan, M. V., Uversky, V. N., and Khan, R. H. (7487), 191−6.

F DOI: 10.1021/acschemneuro.8b00546
ACS Chem. Neurosci. XXXX, XXX, XXX−XXX
ACS Chemical Neuroscience Review

(33) Thompson, A. A., Liu, W., Chun, E., Katritch, V., Wu, H., stimulant and convulsant effects of cocaine by 17-substituted-3-
Vardy, E., Huang, X. P., Trapella, C., Guerrini, R., Calo, G., Roth, B. hydroxy and 3-alkoxy derivatives of dextromethorphan. Pharmacol.,
L., Cherezov, V., and Stevens, R. C. (2012) Structure of the Biochem. Behav. 74 (2), 313−323.
nociceptin/orphanin FQ receptor in complex with a peptide mimetic. (54) Choi, D. W. (1987) Dextrorphan and dextromethorphan
Nature 485 (7398), 395−9. attenuate glutamate neurotoxicity. Brain Res. 403 (2), 333−336.
(34) Felden, L., Walter, C., Harder, S., Treede, R. D., Kayser, H., (55) Zhang, W., Shin, E.-J., Wang, T., Lee, P. H., Pang, H., Wie, M.-
Drover, D., Geisslinger, G., and Lotsch, J. (2011) Comparative clinical B., Kim, W.-K., Kim, S.-J., Huang, W.-H., Wang, Y., et al. (2006) 3-
effects of hydromorphone and morphine: a meta-analysis. Br. J. Hydroxymorphinan, a metabolite of dextromethorphan, protects
Anaesth. 107 (3), 319−28. nigrostriatal pathway against MPTP-elicited damage both in vivo
(35) Armstrong, S. C., and Cozza, K. L. (2003) Pharmacokinetic and in vitro. FASEB J. 20 (14), 2496−2511.
drug interactions of morphine, codeine, and their derivatives: theory (56) Zhang, W., Qin, L., Wang, T., Wei, S.-J., Gao, H.-m., Liu, J.,
and clinical reality, Part II. Psychosomatics 44 (6), 515−20. Wilson, B., Liu, B., Zhang, W., Kim, H.-C., and Hong, J.-S. (2005) 3-
(36) Inturrisi, C. E. (2002) Clinical pharmacology of opioids for hydroxymorphinan is neurotrophic to dopaminergic neurons and is
pain. Clin J. Pain 18 (4 Suppl), S3−13. also neuroprotective against LPS-induced neurotoxicity. FASEB J. 19
(37) Wermeling, D. P. (2015) Review of naloxone safety for opioid (3), 395−397.
overdose: practical considerations for new technology and expanded (57) Snyder, E. W., Shearer, D. E., Beck, E. C., and Dustman, R. E.
public access. Ther. Adv. Drug Saf. 6 (1), 20−31. (1980) Naloxone-induced electrographic seizures in the primate.
(38) Chiang, C. N., and Hawks, R. L. (2003) Pharmacokinetics of Psychopharmacology 67 (3), 211−214.
the combination tablet of buprenorphine and naloxone. Drug Alcohol (58) Somera Molina, K. C., Robin, B., Somera, C. A., Anderson, C.,
Depend. 70 (2 Suppl), S39−47. Stine, C., Koh, S., Behanna, H. A., Van Eldik, L. J., Watterson, D. M.,
(39) Helm, S., Trescot, A. M., Colson, J., Sehgal, N., and Silverman, and Wainwright, M. S. (2007) Glial activation links early-life seizures
S. (2008) Opioid antagonists, partial agonists, and agonists/ and long-term neurologic dysfunction: evidence using a small
antagonists: the role of office-based detoxification. Pain Physician 11 molecule inhibitor of proinflammatory cytokine upregulation.
(2), 225−35. Epilepsia 48 (9), 1785−1800.
(40) Pond, S. M., and Tozer, T. N. (1984) First-pass elimination. (59) Figuera-Losada, M., Rojas, C., and Slusher, B. S. (2014)
Basic concepts and clinical consequences. Clin. Pharmacokinet. 9 (1), Inhibition of microglia activation as a phenotypic assay in early drug
1−25. discovery. J. Biomol. Screening 19 (1), 17−31.
(41) Khroyan, T. V., Wu, J., Polgar, W. E., Cami-Kobeci, G., Fotaki, (60) Taylor, C. P., Traynelis, S. F., Siffert, J., Pope, L. E., and
N., Husbands, S. M., and Toll, L. (2015) BU08073 a buprenorphine Matsumoto, R. R. (2016) Pharmacology of dextromethorphan:
analogue with partial agonist activity at mu-receptors in vitro but Relevance to dextromethorphan/quinidine (Nuedexta®) clinical
long-lasting opioid antagonist activity in vivo in mice. Br. J. Pharmacol. use. Pharmacol. Ther. 164, 170−182.
172 (2), 668−80. (61) Ferkany, J. W., Borosky, S. A., Clissold, D. B., and Pontecorvo,
(42) Elkader, A., and Sproule, B. (2005) Buprenorphine: clinical M. J. (1988) Dextromethorphan inhibits NMDA-induced convul-
pharmacokinetics in the treatment of opioid dependence. Clin. sions. Eur. J. Pharmacol. 151 (1), 151−154.
Pharmacokinet. 44 (7), 661−80. (62) Shin, E.-J., Lee, P. H., Kim, H. J., Nabeshima, T., and Kim, H.-
(43) Threlkeld, T., Parran, T. V., Adelman, C. A., Grey, S. F., and C. (2008) Neuropsychotoxicity of abused drugs: potential of
Yu, Y. (2006) Tramadol versus buprenorphine for the management of dextromethorphan and novel neuroprotective analogs of dextro-
acute heroin withdrawal: a retrospective matched cohort controlled methorphan with improved safety profiles in terms of abuse and
study. Am. J. Addict. 15 (2), 186−191. neuroprotective effects. J. Pharmacol. Sci. 106 (1), 22−27.
(44) Commiskey, S., Fan, L. W., Ho, I. K., and Rockhold, R. W. (63) Vezzani, A., Aronica, E., Mazarati, A., and Pittman, Q. J. (2013)
(2005) Butorphanol: effects of a prototypical agonist-antagonist Epilepsy and brain inflammation. Exp. Neurol. 244, 11−21.
analgesic on kappa-opioid receptors. J. Pharmacol. Sci. 98 (2), 109− (64) Khurgel, M., Switzer Iii, R. C., Teskey, G. C., Spiller, A. E.,
16. Racine, R. J., and Ivy, G. O. (1995) Activation of astrocytes during
(45) Heel, R. C., Brogden, R. N., Speight, T. M., and Avery, G. S. epileptogenesis in the absence of neuronal degeneration. Neurobiol.
(1978) Butorphanol: a review of its pharmacological properties and Dis. 2 (1), 23−35.
therapeutic efficacy. Drugs 16 (6), 473−505. (65) Ravizza, T., Rizzi, M., Perego, C., Richichi, C., Veliskova, J.,
(46) Dayer, P., Collart, L., and Desmeules, J. (1994) The Moshe, S. L., De Simoni, M. G., and Vezzani, A. (2005) Inflammatory
pharmacology of tramadol. Drugs 47 (1), 3−7. response and glia activation in developing rat hippocampus after
(47) Miotto, K., Cho, A. K., Khalil, M. A., Blanco, K., Sasaki, J. D., status epilepticus. Epilepsia 46, 113−117.
and Rawson, R. (2017) Trends in tramadol: pharmacology, (66) Marchi, N., Granata, T., and Janigro, D. (2014) Inflammatory
metabolism, and misuse. Anesth. Analg. 124 (1), 44−51. pathways of seizure disorders. Trends Neurosci. 37 (2), 55−65.
(48) Singh Balhara, Y. P., Parmar, A., and Sarkar, S. (2018) Use of (67) Rizzi, M., Perego, C., Aliprandi, M., Richichi, C., Ravizza, T.,
tramadol for management of opioid use disorders: Rationale and Colella, D., Veliskova, J., Moshe, S. L., De Simoni, M. G., and Vezzani,
recommendations. J. Neurosci. Rural Pract. 9 (3), 397. A. (2003) Glia activation and cytokine increase in rat hippocampus by
(49) Grond, S., and Sablotzki, A. (2004) Clinical pharmacology of kainic acid-induced status epilepticus during postnatal development.
tramadol. Clin. Pharmacokinet. 43 (13), 879−923. Neurobiol. Dis. 14 (3), 494−503.
(50) Dunn, K. E., Tompkins, D. A., Bigelow, G. E., and Strain, E. C. (68) Tufekci, K. U., Genc, S., and Genc, K. (2011) The endotoxin-
(2017) Efficacy of tramadol extended-release for opioid withdrawal: a induced neuroinflammation model of Parkinson’s disease. Parkinson’s
randomized clinical trial. JAMA psychiatry 74 (9), 885−893. Dis. 2011, 1−25.
(51) Kim, H.-C., Nabeshima, T., Jhoo, W.-K., Ko, K. H., Kim, W.-K., (69) Smith, J. A., Das, A., Ray, S. K., and Banik, N. L. (2012) Role of
Shin, E.-J., Cho, M., and Lee, P. H. (2001) Anticonvulsant effects of pro-inflammatory cytokines released from microglia in neuro-
new morphinan derivatives. Bioorg. Med. Chem. Lett. 11 (13), 1651− degenerative diseases. Brain Res. Bull. 87 (1), 10−20.
1654. (70) Hu, S., Sheng, W. S., Lokensgard, J. R., and Peterson, P. K.
(52) Tortella, F. C., and Musacchio, J. M. (1986) Dextro- (2002) Morphine induces apoptosis of human microglia and neurons.
methorphan and carbetapentane: centrally acting non-opioid Neuropharmacology 42 (6), 829−836.
antitussive agents with novel anticonvulsant properties. Brain Res. (71) Svensson, A.-L., Bucht, N., Hallberg, M., and Nyberg, F. (2008)
383 (1−2), 314−318. Reversal of opiate-induced apoptosis by human recombinant growth
(53) Zapata, A., Gasior, M., Geter-Douglass, B., Tortella, F. C., hormone in murine foetus primary hippocampal neuronal cell
Newman, A. H., and Witkin, J. M. (2003) Attenuation of the cultures. Proc. Natl. Acad. Sci. U. S. A. 105, 7304.

G DOI: 10.1021/acschemneuro.8b00546
ACS Chem. Neurosci. XXXX, XXX, XXX−XXX
ACS Chemical Neuroscience Review

(72) Pal, A., and Das, S. (2013) Chronic morphine exposure and its
abstinence alters dendritic spine morphology and upregulates Shank1.
Neurochem. Int. 62 (7), 956−964.
(73) Carliss, R. D., Radovsky, A., Chengelis, C. P., O'Neill, T. P., and
Shuey, D. L. (2007) Oral administration of dextromethorphan does
not produce neuronal vacuolation in the rat brain. NeuroToxicology 28
(4), 813−818.

H DOI: 10.1021/acschemneuro.8b00546
ACS Chem. Neurosci. XXXX, XXX, XXX−XXX

View publication stats

Das könnte Ihnen auch gefallen