Agmatine

Last Updated: October 31, 2023

Agmatine is a metabolite of L-Arginine. It shows promise for alleviating neuropathic pain and drug addiction and shows some potential in protecting against strokes and benefitting cognitive health.

Agmatine is most often used for.



Don't miss out on the latest research

1.

Sources and Structure

1.1

Sources

Agmatine is a molecule (specifically, a biogenic amine,[1]) which is a decarboxylated form of L-Arginine and has the molecular name of 4-(aminobutyl)guanidine;[2] prior to its naming as Agmatine, it was referred to as 'Clonidine-displacing substance' as it was discovered to displace bound Clonidine from receptors.[3]

Agmatine is a small biogenic amine that can serve as a signalling molecule in the human body; it is a decarboxylated form of the amino acid L-arginine

Agmatine is a bacterial byproduct (due to expression of the enzyme that makes agmatine from arginine being expressed in bacteria) and as such it is found in fermented foods including:[4]

  • Wine at up to 6.5ppm in white and 22ppm in red[5][6]
  • Beer at 0.5–42ppm[7][8][9]
  • Sake at 114ppm[10]
  • Coffee (instant) at 0.4–5.3ppm[11]
  • Terrestrial meat products (3.1ppm normally, increased to 27ppm after cooking and up to 42ppm if fermented)[12][13][14][15]
  • Fish (highly variable at undetectable up to 401ppm)[16][17][18]

Food products that have failed to have a detectable agmatine concentration include green and roasted coffee,[19] flour,[20] and both soy sauce and cabbage juice.[21]

Agmatine is found in a variety of food products alongside other polyamines, but the concentrations found in food is significantly lower than standard supplemental dosages and likely not biologically relevant

1.2

Structure and Properties

Agmatine is a biogenic amine with one or two positive charges, which prevents it from passively transporting across membranes (requiring transporters).[22] The molecule is a strong base (alkaline) normally,[23] is diprotonated form dominates at a pH of 7.2 (near physiological concentrations) and is the most stable (1325kJ/mol) followed by the monoprotonated (1070kJ/mol) and neutral (705kJ/mol)[24] with a pKa of 8.93.[25]

Due to its lipophilicity and high level of hydrogen bonds, it is hypothesized to be poorly absorbed.[25]

image

1.3

Biosythesis

The enzyme expression of Arginine Decarboxylase (ADC) in human tissues[26] appears to differ from bodily areas where Agmatine naturally accumulates,[27][28] with this discord through to be due to most bodily Agmatine not being due to biosynthesis via ADC.[2]

In general, the mRNA of the ADC enzyme is relatively highly expressed in the liver and kidneys with detectable levels in skeletal muscle, the small intestine,[29][30][31][32] and brain (higher in hypothalamus, medulla oblongata, and hippocampus; comparatively lower in the striatum and cerebral cortex with the PVN being noted.[33] Mostly on neurons, with the highest immunohistology appearing on neocortical and hippocampal interneurons[33]); some in immune cells such as macrophages[34] and lymphocytes.[35]

When looking at where Agmatine accumulates in the body, rat studies suggest that the highest levels appear to be in the stomach, small intestine and adrenal gland[27][28] with some presence in smooth muscle and endothelial cells[36] with comparatively lower concentrations detected in heart, spleen, aortic and brain tissue.[27][28] When looking at areas of the brain that are comparatively higher in Agmatine, the rat hypothalamic paraventricular and supraoptic nuclei appear to have high concentrations[37] usually in terminals of neurons forming excitatory synapses with pyramidal neurons.[38] Comparatively concentrated areas in the lower brainstem (visceral relay nuclei the nucleus tractus solitarii and pontine parabrachial complex, and periventricular areas including the laterodorsal nucleus, locus coeruleus and dorsal raphe[39]), midbrain (ventral tegmental area and periaqueductal gray[39]), and forebrain (preoptic area, amygdala, septum, bed nucleus of the stria terminalis, midline thalamus, and the hypothalamus[39]) have been reported. A few sources suggested that not only neurons have Agmatine, and that astrocytes[40] and the chromaffin cells of the adrenal medulla[41] have expressed Agmatine concentrations.

Four other sources of Agmatine are currently known including gut microflora and dietary sources.[42] Enterohepatic circulation may also account for preserving Agmatine concentrations in the body.[42][2]

1.4

Metabolism

Although L-Arginine directly converts into nitric oxide (via conversion into L-citrulline and giving off NO as a byproduct after being subject to the nitric oxide synthase enzyme), agmatine is unable to be a substrate for nitric oxide.[43][44][45][46] Although agmatine may influence nitric oxide metabolism, it is not due to being converted into nitric oxide.

Agmatine is not a metabolic precursor of nitric oxide

One possible pathway of metabolism of agmatine is being subject to the diamine oxidase (DAO) enzyme,[3] which also handles metabolism of histamine and is known as histaminase,[47][48] and when agmatine is subject to DAO, the product is 4-guanidinobutyrate.[49]

This pathway has been suggest to account for up to 50% of agmatine metabolism (in rat hepatocytes),[49] and inhibiting this enzyme approximately doubles plasma agmatine in rats (0.7 to 1.3µM in this study).[47] DAO is expressed in high levels in kidney tissue, gastrointestinal and epithelial tissue, and smooth muscle cells of the endothelium yet does not appear to be present in skeletal or cardiac muscle, liver, brain, or adrenals.[47]

Agmatine can convert to 4-guanidinobutyrate via the DAO enzyme, which is possibly the main catabolite of agmatine. This enzyme appears to be expressed in high levels in mucus membranes, kidneys, and endothelium

Agmatine is able to convert into the polyamine putrescine, and can do this directly via the agmatinase enzyme, which gives off urea as a byproduct[50][43] or indirectly via conversion into carbamoyl putrescine (agmatine deiminase enzyme), which gives off ammonia as a byproduct, and is then converted into putrescine itself (carbamoyl phosphate as byproduct) via the putrescine transcarbomylase enzyme.[51] Regardless of the pathway, around 10% of agmatine appears to be metabolized into polyamines.[49]

Agmatine has been noted to increase activity of the protein known as antizyme,[52] which can suppress intracellular accumulation of polyamines[52] and polyamine synthesis from ornithine via inhibition of L-ornithine decarboxylase;[53] this enzyme is induced by polyamines themselves, and agmatine is the only known molecule to induce this enzyme that is not a polyamine.[54]

Agmatine is an intermediate in polyamine synthesis between arginine and the polyamine known as putrescine, and the conversion of agmatine into putrescine can take one of two pathways to produce either urea or ammonia. Despite this, however, agmatine may be inhibitory on polyamine bioactivity by reducing the amount of polyamines in cells

One in vitro study has also noted that approximately 1-3% of agmatine incubated in rat hepatocytes was converted into GABA,[49] which is much less than polyamines (10%) and guanidinobutyraldehyde (50%); 30% of agmatine was not metabolized in this study.

Likely not practically relevant

If the latter pathway is chosen and carbamoyl phosphate is produced, it can be metabolized into carbamate via the carbamate kinase enzyme, which simultaneously converts ADP into ATP. This pathway, known as the deiminase pathway, is a way for bacteria to produce ATP from arginine and agmatine.[55][56][57]

The above mechanism is common to transcarbomylase enzymes (that produce carbamoyl phosphate), as both ornithine transcarbomylase[58][59] and oxamate transcarbamylase[60][61] have been linked to bacterial ATP production through carbamate production.

In bacteria at least (human relevance not known and probably doubtful), agmatine can be used to create ATP

1.5

Regulation

Serum concentrations of agmatine have been detected in the ranges of 6.8+/-0.6ng/mL[62] and 6.8-16.9ng/mL[63] in rats, but appear to be significantly higher in humans such as 47ng/mL,[64] 33.8+/-16.6ng/mL,[65] or 79.42-82.44ng/mL.[66]

Cerebrospinal fluid (CSF) concentrations of agmatine in rats are in the range of 6.1-23.5ng/mL,[63] while those in humans range from 24.3-54.0ng/mL.[65] The latter study noted a possible correlation between CSF and serum agmatine concentrations.[65]

Neural concentrations in rats have been noted to be in the range of 15.3+/-2.4ng/g,[62] while bovine brain has higher concentrations (1.5-3.0nmol/g or 0.2-0.4µg/g).[67] The concentrations in the bovine brain are correlated with those of catecholamines, as 0.2-0.4µg/g of agmatine in cow brain is similar to 0.5µg/g concentrations seen for both noradrenaline and dopamine.[67]

Agmatine has a regulated serum concentration and it appears to be in the range of 50ng/mL or higher for humans than it is for rats. Its concentrations are similar to those seen with catecholamines

Plasma agmatine levels (independent of supplementation) have been noted to be significantly reduced in metabolic syndrome (by 3.7%)[66] and significantly elevated in both schizophrenia[68] and depression.[69]

Agmatine appears to be reduced in metabolic syndrome (by a small amount) and elevated in some neurological disorders such as depression and schizophrenia

2.

Pharmacology

2.1

Transportation in Serum

Agmatine has been found to be absorbed following oral ingestion[70] and oral ingestion appears to influence cognition[71] (suggesting that it can reach the brain).

When looking at the distrubution, low doses (9nmol in rats) is able to increase bodily concentrations of agmatine in all tested organs (stomach, intestines, liver, spleen, lungs, brain, colon, kidneys, adrenals, heart, and skeletal muscle) with 64+/-7% being detected in the liver after 3 hours.[70]

Appears to be absorbed following oral ingestion and readily distributed to tissues; it appears to reach the brain following oral ingestion

In systemic circulation, agmatine appears to have a half-life of less than 10 minutes[72][73] although in the brain it appears to have a half-life exceeding 12 hours.[72]

There appears to be a fairly short half-life in systemic circulation (not the brain), while the half-life in the brain itself appears to be much longer

2.2

Cellular Uptake

Agmatine cannot passively cross plasma membranes due to being protonated at physiological pH, and thus requires transporters. It does appear to be readily taken up by rat hepatocytes (rate of 0.37+/-0.04nmol/h/mg protein).[49] Uptake into cells appears to have some overlap with putrescine transportation, as high concentrations of putrescine can inhibit agmatine uptake[70][74] and vice versa.[49] Transportation is energy dependent.[74]

The Extraneuronal Monoamine Transporter (EMT) and Organic Cation Transporter 2 (OCT2) transporters can accept agmatine with similar capacities, with the OCT1 transporter being significantly (9-fold) less active.[75] These receptors are saturatable at 1-2mM (Km) with a Vmax of 8-16nmol/min/mg protein (11.5+/-1.1 for OCT2 and 15.9+/-3.5 for EMT).[75]

Agmatine appears to be taken up by transporters into cells and tissue, and the transporters used have overlap with putrescine transporters. EMT and OCT2 are suspected of being the transporters

2.3

Neurological Distribution

When looking at specific brain regions, agmatine in basal conditions (without supplementation) has been detected in the brains of rats in the cortex (14.9-16.6ng/g), hypothalamus (19.5-23.9ng/g), medulla (22.2-25.9ng/g), cerebellum (20.6-37.0ng/g), and hippocampus (23.6-33.1ng/g).[62] An overall immunohistological approach notes that most agmatine concentration is present in the cerebral cortex and subiculum (inferior portion of the hippocampus), but can be detected in other brain regions when an axon transport inhibitor is used (such as the forebrain and brainstem); there is no detectable agmatine containined neurons in the cerebellum nor spinal cord.[39]

Agmatine concentrations in the brain are correlated with imidazoline receptor concentrations[64] and with concentrations of the enzyme that creates agmatine (arginine decarboxylase);[76][67] the enzyme that degrades agmatine (agmatinase) appears to also be well expressed in the hypothalamus and hippocampus but not in the cortex.[50]

Agmatine is expressed in various brain regions, and is particularly rich in the inferior portion of the hippocampus and the cerebral cortex

Agmatine is known to cross the blood brain barrier where 10, 50, and 300mg/kg injections of agmatine into mice noted that the highest dose was able to increase neural agmatine to approximately 800ng/g (700% higher than baseline) and in monkeys injected with 25-200mg/kg agmatine it was found that the rise in cerebral agmatine (11.3μM or 1,469ng/mL) was approximately 16% that of the plasma rise in agmatine (70.2μM or 9,126ng/mL).[63] There appears to be a rather long elimination phase from the brain (between 24-72 hours).[77]

In both mice and monkeys, L-arginine injections failed to increase brain agmatine.[63]

Agmatine can increase brain agmatine concentrations at high doses, and can cross the blood brain barrier. Plasma agmatine rises to a larger degree than does neural agmatine concentrations, and administering L-arginine does not increase agmatine concentrations

3.

Neurology

Agmatine is currently considered to be a relatively new neurotransmitter, since it has its metabolism (both creation[78] and destruction[79]) regulated in neurons and is stored presynaptically in these neurons;[39][80] following transport to nerve terminals,[81][82][80][83] agmatine is released upon depolarization of the neuron, which is mandatory for neurotransmitters.[80][83] Agmatine can also be taken up by and stored in astrocytes.[84]

Agmatine is also sometimes referred to as a neuromodulator. This is likely related to the observations that agmatine can positively modulate signalling through the NMDA receptor (via putrescine[85]), α2 adrenergic receptors,[86] and serotonergic receptors (5-HT1A/1B)[87] at low concentrations, while inhibiting binding at the polyamine binding site[88] (that putrescine acts via binding to[89]), competitively inhibiting the α2 adrenergic receptors,[86] and being antagonistic with 5-HT1A/1B signalling[90] at higher concentrations.

Agmatine is a novel neurotransmitter substance, and due to this has a large amount of mechanisms and interactions with other neurotransmitter systems. It is sometimes referred to as a neuromodulator as well, due to some of its mechanisms being bidirectional depending on contexxt

3.1

Imidazoline Neurotransmission

Agmatine is a ligand for imidazoline receptors with most affinity towards the I1 receptor subset followed by I2b.[91] Its affinity is greater enough to displace idazoxan from the receptor, similar to how it can displace clonidine from the α2A receptor[92] and has been calculated (EC50 values) at 0.7µM and 1µM for I1 and I2 receptors, respectively.[67]

Agmatine is an Imidazoline receptor agonist (activator) with fairly high affinity

Downstream of the imidazoline receptor lays an increase in β-endorphin secretion.[93]

Activation of imidazoline receptors underlies an increase in endorphins (and the effects of that, likely analgesia and blood glucose reductions)

3.2

Adrenergic Neurotransmission

Agmatine is known to interact with the alpha-2 (α2) subsets of the adrenergic receptors, and has affinity for all four subsets (variable Ki between 0.8–164μM[94][86][67][95][94] and an EC50 of 4µM has been noted to be lower than noradrenaline at 0.8µM, the main ligand of adrenergic receptors,[67]) while having poor or no significant affinity for the alpha-1 (α1) subset nor β-adrenergic receptors.[86][67] In regards to its affinity for the α2A receptors, it can displace clonidine from the binding site.[91][67]

One study has noted that 10µM agmatine has shifted the concentration-response curve to the left (indicative of enhancing the signalling of noradrenaline and the agonist moxonidine with IC50 values of 7.76 and 6.86), while 100-1,000µM agmatine had the opposing effect and hindered signalling; the authors concluded that at low doses agmatine is a positive allosteric modulator of α2A signalling while at higher concentrations it inhibits signalling competitively.[86]

Agmatine has high affinity for the alpha-2 adrenergic receptors, with little to no affinity for the alpha-1 subsets nor the entire class of beta-adrenergic receptors. It appears to be a positive allosteric modulatory at low concentrations (enhancing the signals of other ligands without inherently activating the receptor), while at higher concentrations it can act as a competitive inhibitor

Numerous studies in vitro have failed to find a per se agonistic property of agmatine on these receptors (in line with the role of an allosteric modulator, which does not inhernetly act on the signalling of a receptor),[96][95] while those in living models have noted that some effects of agmatine can be abolished with α2A receptor inhibitors (yohimbine and rauwolscine most commonly).[97][98]

Agmatine definitely appears to signal through α2A receptors in living models, possibly due to the allosteric modification mentioned earlier. This is not always seen in vitro, possibly due to not introducing other ligands into the medium (as agmatine would enhance the signalling of other ligands, it presumes another ligand is present)

Agmatine has its concentrations correlated with catecholamines in the bovine brain (0.2-0.4µg/g for agmatine, 0.5µg/g for dopamine and noradrenaline)[67] and is known to influence a subset of catecholamine receptors (the α2 adrenergic receptors) while not significantly influencing the others (α1 adrenergic, β-adrenergic, and dopamine D2 receptors).[67]

Agmatine is thought to interact with monoamine oxidase (MAO) enzymes as imidazoline ligands usually have this property since MAO enzymes are structurally similar to imidazoline receptors.[99][100] Agmatine has been found to be a ligand for MAOA near the flavin site (somewhat weak in preventing kynuramine from being oxidized, IC50 at 1,000µM[24]) and elsewhere has been noted to have an IC50 value of 168µM but not be significantly active in vivo.[100]

Agmatine has its concentrations correlated with catecholamine levels, and is known to interact with the MAO enzymes as well although weakly (MAO inhibition is likely not a practical concern with supplemental agmatine due to the high concentration needed)

It shows positive regulation such as a concentration-dependent release of adrenaline and noradrenaline from adrenal chromaffin cells with an EC50 of 5µM[67] (these cells express imidazoline but not α2A receptors,[101] so this was thought to be indicative of acting on imidazoline receptors which induce catecholamine synthesis[102]) and elsewhere has been noted to increase the actions of noradrenaline secondary to α2A receptors (in endothelial cells).[103] This potentiation is cocaine-sensitive (prevented by cocaine, an inhibitor of the noradrenaline transporter) yet agmatine does not modify the noradrenaline transporter per se.[103]

However, activation of the α2A receptors in endothelial cells has been shown to suppress synthesis of catecholamines,[103] while imidazoline receptor activation may also suppress catecholamines.[104]

Agmatine appears to have a regulatory role for catecholamines, and has been implicated in both inducing and suppressing their release. Practical implications of supplemental agmatine are not currently known

3.3

Glutamatergic Neurotransmission

Agmatine has affinity for the NMDA receptors (IC50 value in the 100-300μM range[105]) as the guanidine group of agmatine interacts with the NMDA channel pore,[106] and has been noted to bind to both the MK-801 binding site[107] and the polyamine binding site (Ki of 15μM)[88] but not the glutamate nor glycine binding sites.[105] As agmatine does not have the ability to activate the receptor up to 100μM[106] and can displace MK-801, which is a noncompetitive antagonist and channel blocker of the NMDA receptor.[108] Though there is evidence for inhibition of the receptor and protection from neurotoxic stressors that signal via the NMDA receptor[109][109][109][71] but those that act on the same signalling pathway but circumvent the receptor (calcimycin and staurosporin) are unaffected.[109]

Kainate and AMPA receptors are mostly unaffected, with 3,000μM concentration only inhibiting 15-20% of activity,[106] and four heterodimers (ε1ζ1 through ε4ζ1) are equally inhibited with agmatine.[105]

The binding of agmatine to the polyamine binding site[88] is able to inhibit NMDA signalling from other polyamines such as spermidine from signalling through the receptor[106] via competitive inhibition.[88] This may also extend to histamine, which is thought to bind to the polyamine binding site[110] (not fully confirmed, but may be a unique site[111]) which agmatine can antagonize.[112]

Agmatine binds to two sites of the NMDA receptor (neither of which bind glutamate) and functions as a noncompetitive inhibitor. It can competitively inhibit polyamines from activating the receptor, and the inhibition of NMDA receptors does not extend to the other two glutaminergic receptors (AMPA and kainate)

Due to the role of agmatine as an NMDA inhibitor, it shows protective effects against glutamate excitotoxicity (19-51% reduction of cytotoxicity with 10-1,000µM[109]) and NMDA excitotoxicity with similar potency (46% at 1,000µM)[109].

Lower concentrations of agmatine (1-10µM agmatine) have been found to be bioactive, attenuating the reduction in catecholamines seen during glutamate excitotoxicity[71]

These protective effects in vitro with 1,000µM are comparable to 1,000µM desipramine[71] and to 20µM MK-801.[109]

Inhibition of NMDA appears to be present in vivo following intraspinal or intrathecal injections of low dose agmatine[113][111]

Appears to be neuroprotective against excitotoxicity (from glutamate and NMDA) likely due to blocking the receptor that they signal through, the NMDA receptor. This has been noted in living systems following standard dosing, and although its maximal potency is at a very high concentration likely not attainable with supplementation it does appear to be active at even physiological concentrations

The interactions of agmatine with NMDA receptors in an inhibitory manner is thought to play roles alcohol[114][115] and cocaine dependence,[116] and general neuroprotection against excitotoxicity mediated via the NMDA receptor. Some anti-depressive effects (ultimately influencing potassium and calcium signalling) may also be tied back into NMDA receptor antagonism.

NMDA inhibition appears to be sought after mostly for its antiaddictive and neuroprotective properties, with the latter referring mostly to excitatory toxins (such as excessive glutamate or stimulant usage)

3.4

Nitrergic Neurotransmission

Nitric oxide is covered more in depth in the cardiovascular health section, due to its interactions with blood flow and cardiovascular health. This section solely discusses neurology

Agmatine has been noted to be a neuronal nitric oxide synthase (nNOS) inactivator with a Ki of 29µM secondary to increasing the activity of the NADPH oxidase subunit.[117] L-Arginine is known to hinder the activity of this subunit[118][119] and has been noted to reduce the inactivation of nNOS induced by agmatine.[117]

Inactivation of nNOS either directly (or indirectly via NMDA inhibition as NMDA activation causes an increase in nNOS activity[120]) is thought to underlie the interactions of agmatine and opioids, since NOS inhibitors tend to attenuate tolerance to opioids[121][122][123] and NMDA inhibitors have been implicated as well.[124][123]

Agmatine is able to inhibit the activation of the neuronal isoform of the nitric oxide synthase enzyme (nNOS) in many instances, either directly via inactivation or secondary to the NMDA receptor being inhibited. For any neurological phenomena that involves nNOS suppression, supplemental L-arginine is possible antagonistic

3.5

Cholinergic Neurotransmission

1mM agmatine has been found to inhibit the nicotinic acetylcholine receptor's response to the agonist dimethylphenylpiperazinium (DMPP) by 67+/-11% (showing signs of both competitive and non-competitive inhibition)[125]

May be an antagonist of nicotinic acetylcholine receptors, but needs to be investigated more to confirm

3.6

Serotonergic Neurotransmission

Agmatine does not appear to interact with serotonin per se too much, as it does not influence serotonin release from PC12 neurons (1-100µM)[71] and when serotonin is depleted (70%) the antidepressant effects of agmatine are unaffected.[126] That being said, the antidepressant effects of agmatine appear to be dependent on serotonin receptors as blocking either the 5-HT1A,[90] 5-HT1B,[87] 5-HT2A,[87][90] or 5-HT2C[87] serotonin receptors inhibits antidepressant effects.

Blocking the 5-HT3 receptor does not appear to abolish the antidepressant effects of agmatine[90] perhaps since it has been implicated for being an antagonist itself,[38][127] and agmatine does not appear to be a direct ligand for the 5-HT2 receptors.[67]

Agmatine appears to enhance signalling through the serotonergic receptor, but does not appear to modify synaptic (extracellular) serotonin concentrations nor is it really dependent on them. This augmentation of serotonergic signalling appears to underlie antidepressant effects

Due to the above mechanisms and due to synergism between agmatine and serotonergic antidepressants,[128][98] it is thought that agmatine enhances serotonin signalling. It is not known whether this is due to postsynaptic protein modifications or allosteric modification of the receptor (it appears to be independent of influencing synaptic serotonin concentrations) and it is unclear exactly what mediates these effects as NMDA antagonism[129] and NOS inhibition[130] are both implicated in enhancing serotonergic signalling, while inhibition of both the imidazoline[131][128] and α2A receptors[98] prevent antidepressant effects.

It is unclear how agmatine enhances serotonergic signalling, but signalling through one of the four classical pathways of agmatine (or, likely, some combination of them) appears to then beneficially modify serotonin signalling

3.7

Cannabidergic Neurotransmission

Endocannabinoids are naturally occurring ligands of the cannabinoid receptors (CB1 and CB2; the former more involved with neurology) and are named after the first discovered ligand, THC from marijuana. The CB1 receptors are expressed in a pattern in the midbrain[132][133] that is very similar to the pattern of imidazoline receptors[134] and agmatine localization,[28] and in cardiac tissue at least they have been found to beneficially influence one another.[135]

While agmatine (50-100mg/kg injections) are ineffective at reducing thermal pain perception (hot plate test in rats), 50mg/kg agmatine was able to synergistically increase the pain killing efficacy of two cannabinoidergic drugs by 300-440% via acting on imidazoline receptors,[136] although the effect is also abolished when CB1 antagonists are used.[136] It seems that signalling through imidazole receptors (seen elsewhere when agmatine augmented cannabinoid induced hypothermia[137][138] ) from agmatine will enhance the actions of the CB1 receptor, but still requires an agonist, and it is unlikely that other mechanisms are at play since yohimbine failed to abolish the synergy (excluding α2A receptors[136]) while NDMA antagonism is known to reduce analgesia from CB1 signalling[139] and NOS inhibition is fairly ineffective.[140][139]

It has been noted elsewhere that activation of imidazoline receptors is able to inhibit binding of the CB1 antagonist SR 141716A from its binding site,[141] which suggests imidazoline-induced allosteric modification of the CB1 receptor.

Via signalling through imidazoline receptors, agmatine appears to enhance the pain killing effects of cannabinoids against heat. There appears to be general synergism between the two, and agmatine's role appears to be an indirect positive modulator (possibly by changing the structure of the receptor to better encourage signalling) of CB1 receptors

Potential unexplored synergism for relieving neuropathic pain, as marijuana is well known to do so

3.8

Opioidergic Neurotransmission

The activation of imidazoline receptors (specifically I2 receptors) in the adrenal glands appears to induce the release of β-endorphin (a naturally produced opioidergic pain killer)[93] which has central and peripheral implications.

Enhancement of opioidergic analgesia[124] and the attenuation of tolerance development[142] have been noted with NMDA antagonists before (possible role of Agmatine) although one study has noted that the prevention of tolerance was reliant on activation of imidazoline receptors.[143]

Agmatine is able to release some opioids inherently by activating imidazoline receptors in the adrenal glands. So in regards to the following information of how agmatine interacts with opioids (which should apply to β-endorphin as well), it is somewhat synergistic with itself in persons with functioning adrenals

The acute analgesic (pain killing) effects of morphine are augmented when coadministered with agmatine,[144] which is mediated by α2A receptors[145] and appears to extend to oxycodone[146] and fentanyl (two other opioidergic drugs similar to morphine).

For acute analgesia, agmatine appears to be synergistic with opioids

Agmatine has been found to potently inhibit tolerance to μ-opioid agonists (Endo-2 and DAMGO-AG) with intrathecal injections of 4nmol[113] which appears to be effective for up to 48 hours following a single dose.[113] This has also been noted in rhesus monkeys given oral agmatine (40-80mg/kg).[147]

Tolerance to opioidergic drugs has been noted to be reduced in research animals.[144][121]

Tolerance to opioids (the reduction of efficacy that comes with prolonged usage) appears to be attenuated when agmatine is coadministered, which would synergistically preserve the analgesic effects of opioids

Adrenergic receptors are involved with opioid receptors, with activation of the α2A receptor[148][149] and inhibition of β-adrenergic receptors being able to attenuate morphine withdrawal symptoms;[150] conversely, yohimbine (inhibitor of α2A) negatively augments withdrawal symptoms.[151][152]

In studies that measure addictive properties (self-administration usually what is investigated) or withdrawal symptoms of opioidergic drugs, agmatine has been noted to reduce self-administration of fentanyl.[153][154]

In regards to conditioned place preference (CPP; a preference for one place over another thought to be a biomarker of addictive behaviour[155]), agmatine cotreatment with morphine is able to augment morphine-induced conditioned place preference.[156]

Agmatine appears to be synergistic with opioid drugs such as morphine or fentanyl, preventing their tolerance and possibly reducing the addictive potential thereof. Agmatine appears to be quite potent at this effect

3.9

Pain and Analgesia

Agmatine appears to be a nonproton ligand for ASIC3[157][158] which is one of six acid sensing ion channels[159] that are usually activated by acidity (protons);[160] ASIC3 is somewhat unique since it is not readily desensitized in response to acidity[161][162] and is present in sensory neurons where its activation is associated with the perception of pain.[163][164]

Agmatine can induce signalling at neutral pH (7.4), and is classified as a nonproton ligand since it does not bind to where protons do on the channel (during acid sensing).[158] This activation was unique to agmatine (not seen with polyamines nor arginine), and agmatine was also found to sensitize receptors to acidity (pH 7.0).[158] However, the authors noted that the low potency paired of signalling paired with low concentrations of agmatine normally may mean that this is not a significant concern under normal conditions[158] although pain can be induced with agmatine in mice via ASIC3 dependent means.[157]

Conditions in which acid-induced pain is relevant (blood pH of around 7.0) include inflammation, infection, ischemia, hematomas, and exercise[159] while conditions where agmatine is elevated (alongside increases in percieved pain) include cancer and trauma patients.[165][34]

Agmatine is able to activate the ASIC3 receptor which is involved in percieving pain associated with acidity, and via this mechanism it is theoretically pain causing. Practical significance of this pathway is not known, but at this moment in time it is plausible that agmatine normally does not signal pain but unnormally high concentrations can augment acid-induced pain

In an acetic-acid writhing test (visceral pain model), agmatine has weak analgesic properties.[143] In tests of inflammatory or neuropathic pain, agmatine produces dose-dependent pain reduction.[166][167]

Agmatine by itself has been found to either have no activity in pain relief[136] or to have a short lasting activity (10-30 minutes, only against thermal hyperalgesia) in rats.[72] The short lasting effects may be related to agmatine's short half-life (less than 10 minutes[72]) and secretion of β-endorphin from the adrenal glands.[93]

Agmatine does not appear to be effective against heat pain and fairly weak against visceral pain. Despite that, agmatine appears to have dose-dependent pain relieving properties on inflammatory and neuropathic pain

One double-blind study (partly funded by Trimarco Medical Division, conducted by patent holders) using 2.67g of Agmatine Sulfate for 14 days in patients with lumbosacral spine degenerative pathologies associated with radiculopathy (n=61) noted that after 14 days there were reductions in pain as assessed by the VAS rating scale, the McGill pain questionnaire, but no significant improvement on the Oswestry Disability Index.[168] These benefits appeared to still be statistically significant after Agmatine sulfate was discontinued for 60-65 days.[168]

Agmatine in isolation appears to be effective in reducing the perception of pain in this particular disease state, and requires more research on it since the degree of pain reduction was remarkable

3.10

Addiction

The α2A receptors are somewhat central to addiction, where its activation is thought to be helpful for addiction (as seen with clonidine and nicotine addiction[169][170]) and its inhibition thought to be a negative influence (augmentor) of addiction (some limited evidence for yohimbine acting in a pro-addictive manner).

α2A receptors are also upregulated in response to addictive drugs such as nicotine[171] which augments stimulation (assessed by locomotion in rats) and addiction with subsequent dosages relative to a single dose.[172] Preventing this upregulation is currently hypothesized to be the mechanism by which agmatine prevents nicotine-induced conditional hyperlocomotion[173] and the increase in locomotion seen over a week of chronic dosing.[172]

Agmatine appears to prevent the increase in hyperlocomotion with chronic nicotine ingestion, which is possibly due to preventing upregulation of α2A receptors (which prevents nicotine from releasing more catecholamines with repeated dosages)

Symptoms of alcohol withdrawal such as wet dog shakes/tremors[174] and anxiety[175] are attenuated with agmatine administration. Although the mechanism for this is not yet known, it is thought to be related to NMDA receptor antagonism since polyamines are known to activate the NDMA receptor in a neurotoxic manner during alcohol withdrawal[176][177] and agmatine is an established inhibitor.

May be useful during alcohol withdrawal, and despite nicotine and opioids being related to α2A receptor upregulation the interactions with alcohol are possibly mediated by another mechanism (NMDA antagonism)

Cocaine addiction has been noted to be aided by NMDA inhibition in vitro[116] (with not so promising evidence in humans[178][179]) and by NOS inhibition in planarians (non-mammalian model of animals with mammalian-like neurotransmission),[180] which are two currently hypothesized links between cocoaine addiction and agmatine.

Coadministration of agmatine in a dose sufficient to prevent fentanyl-induced addictive behaviour has failed to modify cocaine related addictive behaviour in rats.[154] Practical relevance of this data is uncertain since it is known that cocaine is not overly addictive to rats[181] and is lower on the reward ladder than is intense sweetness;[181][182] there is currently no evidence in humans.

Evidence for the anti-addictive properties on cocaine are limited and not overly promising in rat models, but due to known species differences between rats and humans in this regard it is uncertain how agmatine will practically influence cocaine addiction

3.11

Stroke and Ischemia

In general, research animals pretreated with agmatine prior to an experimentally induced stroke show neuroprotection with injections of around 100mg/kg.[183][184][185][186][187][188]

In general, agmatine pretreatment before experimentally induced strokes is highly neuroprotective in research animals and quite reliable as well

Nitric oxide synthase (NOS) enzymes, which agmatine is known to inhibit, differential modulate strokes (iNOS appears to contibute to excessive nitric oxide production and harm[189][190] while eNOS is protective via vasodilation;[191] nNOS is also a bit neurotoxic[192]) and it is thought this is one of the targets of agmatine in stroke prevention, as its inhibition towards iNOS (220μM) is 34-fold more potent than that of eNOS (7500μM)[117] and it has been noted that the typical changes in NOS enzyme content after a stroke (increase in iNOS and acute suppression of eNOS) are attenuated with agmatine pretreatment, with iNOS reduced by 13.35% (6 hours) and 46.30% (24 hours)[193] and normalization appears to occur within 4 days.[183]

Perhaps secondary to reduced iNOS production of nitric oxide (has been confirmed),[188] a reduction in peroxynitrate concentrations have also been detected.[184]

There appears to be beneficial modulation of nitric oxide metabolism in response to agmatine supplementation, where the proinflammatory changes in iNOS are reduced and the protective effects of eNOS preserved. This reduces levels of nitric oxide and peroxynitrate, which in the context of a stroke their abnormal elevations are harmful

The aquaporin-4 receptor is a receptor expressed on astrocytes that selectively uptakes water,[194] and reducing (or abolishing) expression of this receptor enhances survival in brain edema[195] which is induced after ischemia stroke.[196] Agmatine (100mg/kg) has been noted to attenuate the increased expression of aquaporin-4 seen during experimental stroke[185] and possibly secondary to that brain edema from stroke is almost normalized.[183][185]

The expression of aquaporin-4 is significantly reduced with agmatine pretreatment, and due to this less edema occurs following experimental stroke. This is likely an effect of agmatine that is secondary to another target

When looking at neurons and glial cells, astrocytes (glial cell) appear to be preserved with agmatine pretreatment at 100mg/kg injections in vivo[197][183] and in isolated cells (100µM).[186]

The in vitro study confirmed increased NF-kB translocation into the nucleus associated with more activation of IκBα and IKKα/β,[186] which is a cellular protective effect.[198] Although it is not confirmed what mediates this process, agmatine has been noted to activate PI3K/Akt elsewhere[199][200] and this is known to be a cellular survival pathway (Akt signalling via NF-kB to survivin).[201][202][203]

Agmatine appears to protect cells from ischemia/hypoxic death, which may be due to increasing activity of NF-kB which then increases survivin

3.12

Stress and Anxiety

The arginine decarboxylase enzyme has been noted to be induced (increased in activity) by acidity in both plants[204] and bacteria[205] and has been shown to be responsive to osmotic[206] and copper-induced oxidative stress.[207]

Due to the above, and the fact that in mammals subject to cold stress an increase in cerebral (15.3+/-2.4ng/g to 57.4+/-19.6ng/g; 275%[62]) and plasma (6.8+/-0.6ng/mL to 58.1+/-12.8ng/mL; 754%[62]) agmatine is seen, it appears that the arginine decarboxylase enzyme is stress responsive and increases agmatine in periods of stress (although the authors of at least one paper suggest other mechanisms may be at play, since a plasma rise in agmatine was detected in 4 hours which may have been too quick to place causation on the enzyme[62]).

The enzyme that creates agmatine from arginine (known as arginine decarboxylase) appears to be increased under periods of stress, and due to this the stress response is met with a large increase in agmatine. It is thought that this is a protective response to stress

An increase in arginine decarboxylase activity has been noted during hypoxia.[188]

Mechanistically, an intrathecal infusion of agmatine (10-40mcg) can cause concentration dependent increases in the firing of neurons in the locus coeruleus (via nitric oxide signalling[208]). This brain region appears to be involved in the bodily response to stress, particularly anxiety and depression,[209] and thus this is thought to be related to agmatine's actions.

There are a few interactions with alcohol as well, as anxiety caused from alcohol withdrawal is abolished[175] and the anxiety reduction by alcohol is actually agmatine dependent (depleting agmatine prevents it from occurring).[175]

Agmatine is involved in anxiety processing in the brain, and alcohol appears to reduce anxiety via influencing agmatine concentrations

In studies using an elevated maze plus test (EPM; standard anxiety test for rats), 20-40mg/kg injections of agmatine[210] and 40mg/kg[211] have efficacy in reducing anxiety with comparable (nonsignificantly more potent) effects relative to 30mg/kg Imipramine.[211] One study using oral intake of agmatine (10-40mg/kg) noted reduced anxiety in a social conflict test in rats comparable to diazepam, with no effect from 80mg/kg nor 5mg/kg.[212]

In a light-dark test of anxiety, agmatine at similar doses has either failed to have an effect[126] or it was only notable at 80mg/kg injections in mice or 10mg/kg oral intake; both being less potent than the reference drug (diazepam).[212]

100mg/kg has been found to either have no effect[211] or actually worsen anxiety.[210]

Agmatine appears to be able to reduce anxiety at a dosage similar to that where depression is reduced, although there is a demonstratable bell curve with agmatine and double the dose does not have anxiolytic effects. The overall anxiolytic effect at maximal efficacy is either less than or comparable to reference drugs (Valium)

3.13

Depression

Agmatine kinetics (without supplementation) appear to be altered in persons with depression, as there are higher serum concentrations[69] but also higher breakdown as assessed by agmatinase activity in both depressed and bipolar persons.[33] Agmatine is also influenced by pharmaceutical depressants, being reduced in concentration by bupropion (Wellbutrin)[69] while SSRI drugs increase agmatine concentrations.[128] Depression also appears to be associated with perturbed imidazoline receptor signalling.[213]

Depression is associated with altered agmatine kinetics and function, although not really in a predictable manner. Pharmaceutical antidepressants seem to have differing effects on agmatine as well (either decreasing or increasing) despite both being able to treat depression

When used by itself, agmatine appears to reduce immobility time in a forced swim test at 10-40mg/kg oral ingestion (human equivalent being 1.6-6.4mg/kg)[71] which has been confirmed elsewhere with 5-40mg/kg[90] or 10mg/kg oral ingestion[71] and various doses of injections (anywhere between 0.01-100mg/kg; with more efficacy in the 10-50mg/kg range).[98][211][126] For studies using active controls, it was found that agmatine (50mg/kg) is slightly less effective than 15mg/kg imipramine[126] or 10-100mg/kg are all comparable to 30mg/kg imipramine.[211]

Antidepressant effects have also been noted in tail suspension tests[71][98] with 40-80mg/kg oral ingestion being of similar potency as 10mg/kg imipramine.[71]

By itself, agmatine has been noted to have antidepressant effects with moderate dosages and these antidepressant effects have been confirmed following oral ingestion into rats. The potency seems either comparable to or weaker than the reference drug imipramine, but comparisons between the two are not that consistent. The estimated human dose for the rats 10-40mg/kg is 1.6-6.4mg/kg (or 217-435mg for a 150lb person)

Agmatine has been found to work synergistically with numerous antidepressants such as bupropion (at 10-20mg/kg agmatine injections, dependent on imidazoline receptors[131]), imipramine (0.01-50mg/kg,[98] although a failure noted elsewhere[128]), SSRIs (5-10mg/kg agmatine injections, dependent on imidazoline receptors),[128] and oddly putrescine (a polyamine) is synergistic with agmatine on depressive symptoms (0.001mg/kg agmatine injection).[85]

The antidepressant effects are abolished by L-arginine,[98][98] yohimbine (α2A receptor blocker),[90][98][98] and GMP itself;[98] another study noted that potassium channel inhibitors (which appears to be the mechanism that agmatine works via) are inhibtied by PDE5 inhibitors such as Viagra.[214] Blocking either the 5-HT1A,[90] 5-HT1B,[87] 5-HT2A,[87][90] or 5-HT2C[87] serotonin receptors blocks the effects as does blocking the delta and mu subunits of opioid receptors (but not kappa)[215] and opening potassium channels appears to prevent the effects of agmatine.[216]

The effects are not affected by prazosin (an α1A receptor blocker),[98] serotonin depletion,[126] blocking the beta-adrenergic system (propanolol),[87][90] or 5-HT3 receptors.[90]

The inhibition of potassium channels may underlie synergism,[216] as the classical antidepressants mentioned earlier tend to have the ability to inhibit potassium channels[217][218][219][220] as does adenosine (endogenous neurotransmitter)[221] and folic acid[222] while opening potassium channels appears to be a pro-depressant effect.[223][224] It was mentioned earlier that blocking a variety of serotonin receptors inhibits synergism, and serotonin likely mediates potassium channels via its receptor.[225] Finally, despite agmatine not per se inhibiting potassium channels at up to 500µM[226] its antidepressant effects are abolished with potassium channel openers.[216]

The anti-depressant effects of agmatine appear to be via augmenting other agents. While agmatine itself cannot inhibit potassium channels, it (somehow) increases the ability of other agents to inhibit potassium channels which adds to their antidepressant effects. This synergism requires that signalling through serotonin receptors is preserved (which the active molecules likely signal through) and that signalling through the α2A receptors is preserved (which agmatine likely signals through)

One pilot study in human subjects with depression (major depressive disorder and unipolar/bipolar distinctions) and no antidepressant resistance who were given 2-3g agmatine orally without any other pharmaceutical drug noted that the three tested subjects all reported remission of depression[227] which was not reversed by administration of PCPA;[227] suggesting that similar to animal studies[126] agmatine's antidepressive effects are unrelated to serotonin.

One very small pilot study suggests that agmatine is able to induce remission of depression (ie. a curative effect)

3.14

Food Intake

Neuropeptide Y is a peptide neurohormone that positively regulates hunger and due to this is known as an 'orexigenic' (hunger increasing or appetite stimulating) hormone[228] and its injections into the paraventricular nuclei (PVN) of the hypothalamus induces food intake.[229] Neuropeptide Y is highly associated with α2-adrenergic receptors[230][231][232] and their activation (usually via clonidine in studies) increases NPY concentrations;[233][234] this is thought to explain why antagonists (yohimbine and rauwolscine) suppress appetite.[235][236]

Administration of agmatine has been demonstrated to increase food intake in satiated rats, but not hungry rats[237] and infusions of agmatine into the hypothalamus cause increases in food intake (10-20nmol causing 45-54% over 24 hours) associated with increased neuropeptide Y activity,[97] thought to be mediated by α2-adrenergic activation as yohimbine attenuated the effects.[97] Intraperitoneal injections of agmatine (40-80mg/kg) were also effective at increasing food intake (44-49% over 24 hours) while 20mg/kg was ineffective.[97]

Agmatine is somewhat opposite of yohimbine, and via the α2A receptors agmatine infusions can increase appetite, but only in rats that are 'full' rather than already hungry rats. The practical relevance of this information is not known, although promising for those unable to gain weight due to not being able to eat enough

3.15

Memory and Cognition

Agmatine is known to be stored to high levels in the hippocampus, specifically CA1 pyramidal neurons.[38] It is thought to be co-released with glutamate vesicles[238][64] via a synaptic Ca2+-dependent exocytotic process[239] and the levels of agmatine in the synpase increase with hippcampal action potentials (neuronal activation)[239] and the process of learning (60-85% in water maze testing in rats);[240][238][241][240] although higher increases have been reported (210-573%) but only for 85-95 minutes;[242] baseline agmatine does appear to increase, but in the aforementioned lower range.[242] Agmatine appears to attenuate subsequent hippocampal discharges[243]

Agmatine has also been found to be elevated in the stratum radiatum and both prefrontal and perirhinal cortices[244] during the learning process, and is also elevated in the locus coeruleus[39] which has neuronal activity enhanced by agmatine infusions.[208] The locus coeruleus is involved in agmatine's benefits on inhibitory avoidance tasks,[245] the prefrontal cortex is involved in behavioural learning and executive functioning,[246][247] and the perirhinal cortex (as well as the hippocampus) beneficially influence displaced object recognition.[248]

Agmatine's role of a neurotransmitter is involved in memory formation. During learning tasks agmatine is elevated in the hippocampus, prefrontal cortex, stratum radiatum, and perirhinal cortiex and it appears to be a negative regulator of glutaminergic signalling that is coreleased with glutamate (NMDA receptor antagonism likely is involved here as well)

One study (albeit in human mesenchymal stem cells rather than neurons) has noted that increasing activity of the arginine decarboxylase enzyme is able to cause an increase in agmatine (2-fold) which was thought to be the cause for the increase seen in BNDF secondary to phosphorylation of Akt and CREB.[199] BNDF is downstream of CREB[249] which is downstream of Akt[250] which agmatine is known to activate;[200] this pathway is seen as prosurvival in these stem cells[251] but positively regulates synaptic plasticity and memory formation in neurons.[252]

Beyond the possible increase in BDNF (which would play a role in memory formation), oral intake of 20mg/kg agmatine for 2 weeks has increased adenylate cyclase activity in the prefrontal cortex.[90]

Agmatine may have a role in memory formation secondary to enhancing synaptic formation or increasing cAMP activity, the two appear to be regulated by independent means

Conversely, agmatine possesses some amnesiac mechanisms such as antagonism of NMDA receptors[253][254] and the negative regulation on nitric oxide metabolism[255] as they both positively mediate memory formation. Inhibition of NOS has indeed been noted with 40mg/kg injections (as evidenced by a decrease in L-citrulline, indicative of less arginine-citrulline conversion via NOS).[256] Interestingly, an elevation of citrulline is detectable in the dentate gyrus and prefrontal cortex during the learning process.[244]

Although agmatine is co-released with glutamate (agonist of NMDA receptors) under normal non-supplemental conditions, it is not likely a concern; learning tasks increase basal synaptic agmatine from 0.25µM to 0.75µM in the rat brain[242] while the IC50 for inhibiting the NDMA receptor is in the 100-300µM range.[106][105] However, it is plausible that high supplemental levels may inhibit NMDA receptors in the hippocampus.

There are some mechanisms which may be suspicious for being amnesiac, as both NDMA and NOS (which agmatine inhibits) can positively modulate memory formation. Practical relevance of this information is not known

For studies using water-mazes to assess spatial learning, microinjections directly into the hippocampus have failed to augment memory formation[257] and this failure is also seen with intraperitoneal injections of 10-50mg/kg.[258][259][260][261] Agmatine does not appear to modify spatial learning, and one study assessing exploratory learning has found a similar failure.[260]

Contextual fear conditioning and learning has twice been noted to be impaired with agmatine injections (20-40mg/kg intravenously)[258][259] while inhibitory avoidance tasks[245][245] and behavioural learning appear to be enhanced.[260] Displaced object recognition (but not novel object recognition) appears to be enhanced with agmatine infusions as well in the standard dosage range (either 10mcg intrathecal infusion[261] or 40mg/kg intravenous[261]).

Incidentally, the tasks that get benefit with memory formation from agmatine are those where the brain regions normally see pulses of agmatine during the learning process (independent of supplementation). This includes the perirhinal cortex and hippocampus (displaced object recognition)[248] and the prefrontal cortex (behavioural learning),[246][247] whereas the locus coeraleus is known to react to agmatine supplementation and is associated with inhibitory avoidance tasks.[245] The benefits seen with agmatine appear to be both task and delay dependent, and are hypothesized to be associated with more complex rather than simplistic tasks.[262]

Agmatine appears to increase cognition in the brain regions that are eithe responsive to agmatine or are known to release agmatine into the synapse in high concentrations when activated. Due to this agmatine is able to benefit inhibitory avoidance tasks, displaced object recognition, and behavioural learning; it does not appear to influence spatial learning, and may be adverse towards contextual fear conditioning (learning to avoid or fear negative stimuli)

It is likely that there is a bell curve effect and that higher doses are not better, and there is still no evidence that working memory is enhanced with agmatine supplementation.

3.16

Epilepsy and Convulsion

Nitric oxide signalling is known to be a positive mediator of seizures[263] where L-arginine increases seizure susceptability (when other things start them) in a manner attenuated by NOS inhibition.[264] It is thought that agmatine signals via the α2A receptor to reduce nitric oxide production (via downregulating iNOS) to reduce seizure frequency and magnitude.[265]

When a seizure occurs, nitric oxide signalling is enhanced and excessive NO signalling exacerbates the sequelae of the seizure. Agmatine is thought to be protective of seizures by reducing the amount of NO signalling that occurs secondary to acting on its adrenergic receptor, the α2A receptor

Agmatine itself (100mg/kg injection 45 minutes prior to seizure induction) is able to augment phenobarbitol (ED50 reduced from 22.54 to 16.82mg/kg) and valproate (256.1 to 210.6mg/kg) seizure inhibition despite failing to per se protect rats from electroshock (MES) induced seizures[266] and other tested antiepileptics (carbamazepine, lamotrigine, phenytoin, oxcarbazepine, and topiramate) were unaffected by agmatine.[266] Elsewhere, injections of comparable doses (80-160mg/kg, but not lower) were anticonvulsive against MES[267] while lower doses of agmatine (30mg/kg oral ingeston) have been noted to be effective in reducing MES induced seizures within 15 minutes and extending for six hours (most efficacy at four hours[265]) and 60-120mg/kg were not significantly more effective than 30mg/kg.[265]

Lower doses of agmatine (5-40mg/kg; most efficacy from 10-20mg/kg at 30-45 minutes before the seizure) prior to pentylenetetrazole (PTZ) induced seizures have been shown to have anticonvulsive effects[264] in a manner attenuated by yohimbine and nitric oxide induction, but augmented with the NOS inhibitor L-NAME.[264]

Agmatine appears to be active in rodents in suppressing seizures following multiple stimuli (PTZ and MES) and this has been confirmed to occur in rats within 15-360 minutes of acute oral dosing; estimated human equivalent being 5mg/kg bodyweight

3.17

Calcium signalling

Agmatine has a few interactions with calcium signalling. Firstly, NMDA receptor activation tends to increase intracellular calcium which can then activate nNOS[268][269] and via being an NDMA antagonist[106] agmatine can limit the increase in calcium (agmatine can act on nNOS in more than just this way, however). Furthermore, agmatine has been noted to block calcium channels directly on hippocampal neurons in a reversible manner with an IC50 value of 0.79-1.57µM and recorded 21+/-4% inhibition at a concentration of 100nM[226] but elsewhere has been noted to displace the calcium channel blocker known as Diltiazem.[270]

Agmatine has failed to have any influence on sodium or potassium channels at concentrations of up to 500µM.[226] That being said, nitric oxide has been noted to increase the activity of calcium-activated potassium channels[271][272] which is downstream of NMDA activation;[273] this presumes an inhibitory effect of agmatine on potassium channels (as it is an NMDA antagonist), which is thought to at least in part contribute to antidepressant effects.[216]

Overall, it seems that agmatine is antagonistic to calcium signalling (both by preventing NMDA signalling, and by directly inhibiting influx at physiological concentrations) and may indirectly be inhibitory to potassium channel signalling (also via NMDA antagonism)

3.18

Other Mechanisms

Low dose agmatine (5-10mg/kg injections) for 30 days in mice who had memory deficits induced by the diabetic toxin streptozotocin was able to reduce both blood glucose and improve memory[274] although the memory improving effect was independent of the I2 (imidazoline) receptor,[274] which is required for glucose reduction (suggesting two independent mechanisms).[275]

Agmatine has also shown protective effects against memory impairment induced by beta-amyloid pigmentation,[276] the MPTP toxin,[277] scopolamine,[278][279] and inflammation (using lipopolysaccharide to cause inflammation).[280][281]

Agmatine appears to be able to protect the brain from research toxins that induce amnesia and for conditions that may be negative towards memory formation (inflammation and an elevation of blood glucose)

Two studies have noted that agmatine has an ability to disrupt the prepulse inhibition (PPI; a phenomena where a 'prepulse', usually a neuronal action potential, weakly occurs prior to the desired action potential or pulse and due to desensitization attenuates the signal[282]). When PPI is high active action potential are exagerrated and when PPI is low active they are dampened somewhat. Disrupted prepulse inhibition is thought to be related to schizophrenia[283][284] and other neurological ailments such as anxiety and PTSD;[282] agmatine concentrations in the serum of schizophrenia patients is also reliably elevated.[68]

Agmatine injections into research animals have noted that agmatine is able to attenuate PPI in response to sound[285] and the drug phencyclidine.[286] It is additive with apomorphine in this regard,[285] and administration of agmatine to rats is able to attenuate apomorphine induced climbing and amphteamine/ketamine induced hyperlocomotion.[287]

There are interactions with agmatine and prepulse inhibition, a neurological phenomena where a smaller 'prepulse' occurs and suppresses the magnitude of the subsequent 'pulse' due to short-term desensitization

4.

Longevity

4.1

Neurological Aging

During the aging process, there appears to be dysregulation in the nitrenergic (nitric oxide related) system as total NOS activity (not enzyme count) is increased in the prefrontal cortex[288] while agmatine is decreased in the prefrontal cortex and hippocampus (CA1 cells)[289] alongside an increase in polyamines.[290] It is thought that the increase in NOS and decrease in agmatine are related to age-related behavioural impairments.[291][292][293]

Agmatine injections (40mg/kg) daily for 4-6 weeks in aged rats appears to normalize the increased NOS activity in the dentate gyrus and prefrontal cortex without affecting eNOS or nNOS protein content (not significantly affected by the aging process either),[294] which works well with the aformentioned theory as agmatine is also known to correct age-related behavioural impairments at this dose.[260]

Agmatine may be useful in normalizing age-related behavioural deficits

5.

Cardiovascular Health

Agmatine appears to also modulate cardiovascular health in both directions, which is mostly due to interactions with nitric oxide. For the following sections, the positive regulation refers to increasing nitric oxide activity (and vasodilation) while the negative regulation refers to inhibitory effects on nitric oxide activity or vasoconstriction

5.1

Positive Regulatory Mechanisms

Agmatine is a vasorelaxing agent, and at 10µM can increase nitric oxide production in endothelial cells[295] and can increase cGMP concentrations in cells (a normal consequence of nitric oxide acting upon its receptor).[296] This direct stimulation of nitric oxide production (similar to both L-Arginine and D-Arginine) has been found to be secondary to the α2A receptor, and is blocked by rauwolscine at the level of the receptor.[295]. Arginine activates this receptor at 1mM concentration, while the concentration of agmatine required to 100-fold lower.[295]

Mainly due to acting on the α2A receptor, agmatine induces nitric oxide synthesis. This is a similar mechanism to L-arginine (and by extension, L-citrulline) but agmatine appears to be 100-fold more potent at activating this receptor

The overall production of nitric oxide is dependent on G-protein signalling (Gi/o; coupled to the α2A receptor)[295] and NOS enzymes, as inhibitors of NOS enzymes abolish the nitric oxide production.[297] Calcium channel activation[92][295] and ryanodine receptors (RyR) which modulate calcium channels[298] are also involved. The interaction of agmatine and RyRs (which are mediated by nitric oxide) may be due to nitric oxide increasing cyclic ADP-ribose (cADPr) activity;[299][300] cADPr is a positive regulator of the RyR[301][302] and although agmatine does not influence its levels or its binding[303] abolishing the actions of cADPr prevents nitric oxide formation.[303] From there, the increase in calcium can increase activity of eNOS enzymes[304][305] due to activating the protein known as calmodulin.[306]

Increasing nitric oxide via activating the α2A receptor requires that the G-protein coupled to the α2A receptor influences a molecule known as cADPr which then increases the activity of the ryanodine receptor. The ryanodine receptor releases calcium, and this calcium then activates calmodulin which can activate endothelial nitric oxide synthase (eNOS) and subsequent production of nitric oxide

A problem above is that ryanodine receptors (RyRs) are both dependent on and induce nitric oxide formation, which suggest it is both upstream and downstream of NOS enzymes;[307] inhibiting NOS enzymes actually prevents agmatine from acting on RyRs.[303] Interestingly, the effects of agmatine on nitric oxide have also been noted to be impaired by blocking the imidazoline receptors,[92] phospholipase C (PLC),[295] and PI3K[200] which are independent of NOS inhibition (L-NAME has no effect).[296] It is known that there exists signalling from imidazoline[308] and α2A receptors[309] towards PI3K and PLC, there exist a signalling pathway from PI3K via Akt that activates eNOS[310][311] and agmatine has been shown to signal via this pathway.[312]

Activation of the ryanodine receptor actually requires nitric oxide (being both upstream and downstream of NOS enzymes) and this can be rationalized by signalling through both the α2A or imidazoline receptors causing activation of the PI3K/Akt/eNOS pathway, which can also boost production of nitric oxide.

It is plausible that activation of the PI3K/Akt/eNOS pathway causes a short boost in nitric oxide synthesis, while the increased nitric oxide then activates the RyR/Calmodulin pathway to provide a more sustained boost

5.2

Negative Regulatory Mechanisms

There are some pro-contractile properties of agmatine as well. It has been noted that high concentrations of agmatine (1mM) are able to slightly augment the ability of noradrenaline to contract arteries (EC50 value changef from 2.2µM to 1.5µM) while suppressing the procontractile properties of clonidine (0.9µM to 2.6µM) and methoxamine (1.6µM to 2.8µM).[103]

Agmatine may be able to enhance the contractile properties of noradrenaline at high concentrations

There appears to be weak competitive inhibition on all isoforms of NOS with Ki values of 220μM (iNOS), 660μM (nNOS), and 7500μM (eNOS).[117] Other studies have found inhibition of nNOS with an IC50 of 160μM[43] and iNOS with 260μM[313] although the latter study failed to find inhibition of nNOS and eNOS.[313] Other studies have mostly failed to find per se inhibitory effects on NOS enzymes (especially eNOS, which does not appear to be inhibited much if at all)[46][45][44] which is probably due to the high concentration required for inhibition.

Agmatine is able to inhibit the activity of NOS enzymes, but this occurs at fairly high concentrations; while inhibition of iNOS and nNOS are likely not possible without supplementation (but feasible with supplementation) the inhibition of eNOS seems to be irrelevant (too high of a concentration to matter)

Despite the weak competitive inhibition, agmatine is known to be an irreversible nitric oxide synthase (NOS) enzyme inactivator with a Ki of 29μM.[117]

The mechanism appears to be from a 3-fold increase in the activity of the NADPH oxidase subset of NOS (this study used nNOS) which caused a large spike of H2O2 and oxidative inactivation of the enzyme.[117] This was dependent on calmodulin (which transfers an electron from the NADPH oxidase subunit to the heme group of the NOS enzyme; calmodulin itself can reduce NOS activity via inactivation[117]) and after one hour activity was reduced to 30+/-1.2%, omitting NADPH had no effect on the first 20 minutes but significantly inhibited inactivation between 20-60 minutes.[117]

This overall profile of negative regulation (weak competitive inhibitor but an oxidative inactivator) is similar to other NOS inhibitors such as aminoguanidine[314][315] and guanabenz.[316] It is unsure how biologically relevant this is due to the short plasma half-life of agmatine, but unlike the inhibition of the enzyme the inactivation is at a physiologically relevant concentration.

Agmatine can inactivate NOS enzymes, and this occurs at a much more reasonable dosage (three-fold that which activates eNOS). It is not exactly sure how this inactivation affects nitric oxide production in a living system

5.3

Cardiac Tissue

In isolated ventricular myocytes, agmatine (0.1-10mM) has been noted to inhibit calcium uptake into cells[317] and reduce peak amplitude of the L-type calcium channel in a concentration dependent manner.[318]

High concentrations have a calcium blocking effect in isolated cells. Practical relevance of this information is not yet known

5.4

Blood Flow and Coagulation

Vasorelaxation has been noted in kidney cells (causing an increase in glomerular filtration rate)[319][297][303] and cardiac cells (aortic rings)[320][321] which appear to be nitric oxide dependent (hindered by NOS inhibitors and endothelial-dependent).[297]

Agmatine infusions have failed to modify vascular tone in the rat arterial tail vein.[103]

Has some demonstratable relaxing properties in vitro which are dependent on nitric oxide

One study assessing bleeding time (increased by blood thinners) in rats given 300μg/kg agmatine noted an increase in blood pressure following bleeding which improved survival, and that this effect was abolished by yohimbine and arginine (suggesting that it is α2A dependent and relies on a reduction of nitric oxide).[322]

Appears to reduce bleeding associated with an increase in blood pressure, suggesting an overall modulatory role on blood flow rather than a unilateral increase in blood flow

5.5

Blood Pressure

Agmatine is known as clonidine displacing substance since it appears to displace clonidine at both α2A receptors and imidazoline receptors[67] and clonidine is known to act as an antihypertensive (blood pressure reducing) drug via the imidazoline receptor[323][324] which is a property shared by agmatine.[325] Oddly, despite displacing clonidine from the imidazoline receptor agmatine does not interfere with its actions.[73]

Acting on imidazoline receptors is known to reduce blood pressure

In anaesthesized rats, injections of agmatine appear to reduce blood pressure[73][326][327]

6.

Interactions with Glucose Metabolism

6.1

Blood Glucose

Activation of the imidazoline receptors localized in the adrenal gland is known to reduce blood glucose in streptozotocin-induced diabetic rats and agmatine has been found to activate these receptors.[93][275] The reduction of blood glucose appears to be associated with an increase in β-endorphin,[275][93] and β-endorphin is known to increase tissue glucose uptake into skeletal muscle[328][329] independent of whether it is exercised or at rest;[330] possibly due to an increase in GLUT4 mRNA content.[275]

β-endorphin has elsewhere been noted to inhibit hepatic glucose production[331] while inducing insulin secretion from the pancreas;[332] however, it is unsure how agmatine supplementation influences these parameters.

Agmatine can act upon the adrenal glands (via the imidazoline receptors) to release β-endorphin, an opioid that can dispose of glucose from the blood into skeletal muscle tissue. Due to this, agmatine can reduce blood glucose via adrenal gland-dependent means

7.

Skeletal Muscle and Performance

7.1

Mechanisms

Agmatine, secondary to activating imidazoline receptors upon the adrenal glands, can release β-endorphin into the plasma.[275]

β-endorphin (induced by agmatine) is known to increase skeletal muscle glucose uptake in diabetic rats[275][93] independent of whether the muscle is at rest or exercise[330] possibly via GLUT4 mRNA upregulation[275] yet β-endorphin (per se, not induced by agmatine in this study) does not appear to modify adipocyte glucose uptake;[333] this suggest a possible nutrient partitioning effect, but needs to be investigated further to confirm.

Agmatine can reduce blood glucose acutely in diabetic animals, and it is plausible that this is due to a selective disposal of glucose into muscle rather than fat which is due to an increase in β-endorphin. More studies are needed to assess whether or not this actually occurs following oral supplementation

8.

Interactions with Hormones

8.1

Luteinizing Hormone

Injections of agmatine into the hypothalamus appear to dose-dependently cause secretion of luteinizing hormone, thought to be secondary to activating imidazoline receptors.[334]

9.

Interactions with Organ Systems

9.1

Eyes

In vitro, agmatine is able to protect retinal ganglion cells from apoptosis[335] and this has been noted with topically applied agmatine (reducing 55.44% cell death down to 18.65%) which appears to be associated with a reduction in intraocular pressure.[336] This protective effect appears to extend to ischemic ocular injury as well.[337]

Agmatine, applied topically, appears to be protective of the eyes; the implications of oral supplementation is not yet known

9.2

Stomach

Agmatine is normally expressed in stomach tissue at a concentration of around 71ng/g wet weight, which is one of the highest non-brain organ concentrations of agmatine in rats (ahead of the spleen and intestines).[27] It is stored in mucus membranes of the stomach (in parietal cell canaliculi), where its strong alkalinity likely buffers stomach acid from damaging parietal cells.[23]

Agmatine appears to be inhernetly gastroprotective due to its alkalinity, where it protects parietal cells of the stomach from stomach acid

Agmatine at 100mg/kg (injection) has shown protective effects against gastric ischemia (PI3K dependent)[338] but elsewhere has augmented damage on alcohol-induced ulceration[339] and stress-induced ulceration[340][341] via acting on imidazoline receptors in the 20-25mg/kg dosage range; it is not clear if the reason for the difference is due to the dosage or the context of the injury.

α2A receptors appears to induce gastric contractions when activated with imidazoline receptors having no significant role.[342]

Agmatine has been noted to both protect the stomach from damage (against ischemia) yet augment damage done by chemical and stress induced damage

9.3

Intestines

Agmatine is involved with the intestines as it is produced by colonic bacteria (also gastric bacteria[343]) and can accumulated in the intestines following oral administration of agmatine.[70]

9.4

Kidneys

Agmatine is produced within the kidney where it can enhance glomerular filtration rate in kidney tissues possibly via I2 imidazoline receptors (a selective I1 agonist was ineffective)[319] and elsewhere has been found to be dependent on NOS enzymes;[297] the alpha-2-adrenergic receptors do not appear to be required,[319][297] but activation of a class of receptors known as ryanodine receptors (activates calcium signalling) may explain the effects of agmatine.[303] Regardless of the mechanism, it appears that neurons are not required (as innervated and denervated kidneys have similar effects).[319]

Na+/K+ ATPase has also been noted to be increased in renal cells.[344]

Preliminary evidence suggests an increase in glomerular filtration rates, which is supposedly a renoprotective effect. This may be related to a relatively novel mechanism, acting upon ryanodine receptors

9.5

Male Sex Organs

Agmatine is known to interact with the α2A receptors in the vas deferens[95] both presynaptically and postsynaptically[345] where it is able to enhance electricity-induced twitches thought to be associated with blocking potassium channels at 10-1,000µM.[345][346] It was later noted that this enhancement occured in the prostatic portion of the vas deferens but not epididymal portion, and reached maximal enhancement at 1mM (207.1+/-14.2% of control).[346]

10.

Nutrient-Nutrient Interactions

10.1

Arginine/Citrulline

L-Arginine and L-Citrulline are two amino acids that raise plasma arginine concentrations

Agmatine's oxidative inactivation of the nitric oxide synthase enzyme (seen in this study with nNOS with a Ki of 29µM) is due to increasing the activity of the NADPH oxidase subunit[117] which arginine is known to hinder for nNOS[118][119] yet appears to increase activity for iNOS and eNOS.[347][118] In this sense, arginine is antagonistic to the effects of agmatine towards nNOS (a possible reason why supplemental arginine inhibits many neurological effects) but does not affect eNOS in the same manner.

It is not sure whether pairing either of these two agents with arginine will enhance systemic nitric oxide production (via eNOS; the mechanism underlying most of the blood pressure effects) or be antagonistic; both are plausible, with the former being due to increased nitric oxide production from eNOS and the latter due to quicker inactivation of the enzyme.

Arginine (and by extension, citrulline) are inhibitory to some of the neurological effects of agmatine supplementation such as reducing opioid tolerance. The interaction between agmatine and arginine in regards to blood vessels (and 'the pump' for physical exercise) is not clear

10.2

Alcohol

Agmatine is involved with alcohol on both a gastric and neurological level, and overall it can be said that agmatine is good for rehabilitating alcohol withdrawal but probably not the best supplement to coingest.

Alcohol's reduction of anxiety (anxiolysis) is prevented by inhibition of the arginine decarboxylase enzyme, suggesting that it works via agmatine.[175] The increase in anxiety seen during alcohol withdrawal is also normalized with agmatine.[175]

Additionally, the analgesic (pain killing) effects of alcohol appear to be augmented with agmatine injections in rats thought to be due to imidazoline receptor signalling[348] and agmatine has been noted to block the alcohol induced hyperactivity seen in rats (male only) at 5-20mg/kg without affecting locomotion inherently.[349] There does not appear to be any interaction between agmatine and conditioned place preference (CPP), thought to be indicative of no alterations on motivation/addiction.[350]

On a negative side, agmatine is known to be a gastroprotective agent (aids in protecting parietal cells from stomach acid[23]) but has counterintuitively enhanced ulcer formation from alcohol consumption in rats.[339]

At this moment in time, agmatine appears to be useful for alcohol withdrawal. The usage of agmatine alongside alcohol might not be prudent as one study noted an enhancement of stomach ulceration, so timing of agmatine and alcohol consumption would be prudent

10.3

Caffeine

With acute doses of caffeine that are able to induce locomotion (in caffeine naive mice), agmatine at 5-20mg/kg is able to block the increase in locomotion but only in male mice.[351]

10.4

Marijuana

Marijuana is the prototypical cannabinoid drug, which signals through two receptors known as endocannabinoid receptors (CB1 and CB2); the following information should apply to all CB1 agonists (so possibly anandamide from arachidonic acid metabolism and oleamide) with uncertain interactions with CB2 agonists (echinacea).

The CB1 receptors are expressed in a pattern in the midbrain[132][133] that is very similar to the pattern of imidazoline receptors[134] and agmatine localization,[28] and in cardiac tissue at least they have been found to beneficially influence one another.[135]

Endocannabinoid receptors are intimately expressed alongside imidazoline receptors and they appear to positively influence one another; marijuana is a potent activator at CB1 receptors, whereas agmatine is a potent activator of imidazoline receptors

Agmatine has been found to synergistically enhance hypothermia[137][138] and augment the potency of two cannabinoidergic drugs by 300-440% in regards to reducing heat pain (agmatine alone is inactive on this type of pain).[136] This effect is independent of α2A receptors[136] and unlikely to be associated with NOS or NMDA inhibition[140][139] while it was inhibited by both imidazoline antagonists (blockers) and CB1 receptor antagonists.[136] Due to both receptors being required and the ability of imidazoline activation to modify binding of the CB1 receptor antagonist SR141716A to its receptor[141] it is plausible that activation of imidazoline receptors will augment signalling through the CB1 receptor via positive allosteric modification of the receptor (changing the structure of the receptor to modify signalling, positive refers to an increase in signalling relative to baseline).

There appears to be synergism between CB1 receptor signalling and imidazoline receptor signalling. It is not 100% sure how it works right now, but both receptors seem to be important and it is possible that structural changes occur to the CB1 receptor after the imidazoline one is activated (which then enhances signalling). While synergism is only noted for heat pain right now, agmatine has potential to be synergistic with marijuana on a wide variety of mechanisms (most excitingly, neuropathic pain)

11.

Safety and Toxicology

11.1

General

Although the trials cannot be found, an MSDS reports that the acute LD50 of agmatine is 300mg/kg after oral ingestion in mice, 980mg/kg in rats, and 3200mg/kg in rabbits.[352]

A study conducted over 51 days assessing dose tolerability that used 1.335g (10 days), 2.67g (subsequent 10 days), 3.56g (subsequent 10 days followed by 21 days of maintenance) with the dose divided into thrice daily doses with meals in patients with lumbosacral spine degenerative pathologies associated with radiculopathy did not note any significant health effects of supplementation aside from sparse gastrointestinal discomfort when the dose was increased to 3.56g which faded after a few days.[168]

Limited evidence in humans, but currently there are no known side-effects. Insufficient evidence to conclude its level of safety, however

References
1.^Naila A, Flint S, Fletcher G, Bremer P, Meerdink GControl of biogenic amines in food--existing and emerging approachesJ Food Sci.(2010 Sep)
4.^Galgano F, Caruso M, Condelli N, Favati FFocused review: agmatine in fermented foodsFront Microbiol.(2012)
10.^Okamoto A, Sugi E, Koizumi Y, Yanagida F, Udaka SPolyamine content of ordinary foodstuffs and various fermented foodsBiosci Biotechnol Biochem.(1997 Sep)
12.^Ruiz-Capillas C, Jiménez-Colmenero FBiogenic amines in meat and meat productsCrit Rev Food Sci Nutr.(2004)
23.^Steer HThe source of carbon dioxide for gastric acid productionAnat Rec (Hoboken).(2009 Jan)
24.^Jones TZ, Giurato L, Guccione S, Ramsay RRInteractions of imidazoline ligands with the active site of purified monoamine oxidase AFEBS J.(2007 Mar)
26.^Zhu MY, Iyo A, Piletz JE, Regunathan SExpression of human arginine decarboxylase, the biosynthetic enzyme for agmatineBiochim Biophys Acta.(2004 Jan 22)
27.^Raasch W, Regunathan S, Li G, Reis DJAgmatine is widely and unequally distributed in rat organsAnn N Y Acad Sci.(1995 Jul 12)
28.^Raasch W, Regunathan S, Li G, Reis DJAgmatine, the bacterial amine, is widely distributed in mammalian tissuesLife Sci.(1995)
29.^Iyer RK, Kim HK, Tsoa RW, Grody WW, Cederbaum SDCloning and characterization of human agmatinaseMol Genet Metab.(2002 Mar)
30.^Mistry SK, Burwell TJ, Chambers RM, Rudolph-Owen L, Spaltmann F, Cook WJ, Morris SM JrCloning of human agmatinase. An alternate path for polyamine synthesis induced in liver by hepatitis B virusAm J Physiol Gastrointest Liver Physiol.(2002 Feb)
31.^Haenisch B, von Kügelgen I, Bönisch H, Göthert M, Sauerbruch T, Schepke M, Marklein G, Höfling K, Schröder D, Molderings GJRegulatory mechanisms underlying agmatine homeostasis in humansAm J Physiol Gastrointest Liver Physiol.(2008 Nov)
32.^Dallmann K, Junker H, Balabanov S, Zimmermann U, Giebel J, Walther RHuman agmatinase is diminished in the clear cell type of renal cell carcinomaInt J Cancer.(2004 Jan 20)
33.^Bernstein HG, Stich C, Jäger K, Dobrowolny H, Wick M, Steiner J, Veh R, Bogerts B, Laube GAgmatinase, an inactivator of the putative endogenous antidepressant agmatine, is strongly upregulated in hippocampal interneurons of subjects with mood disordersNeuropharmacology.(2012 Jan)
34.^Sastre M, Galea E, Feinstein D, Reis DJ, Regunathan SMetabolism of agmatine in macrophages: modulation by lipopolysaccharide and inhibitory cytokinesBiochem J.(1998 Mar 15)
35.^Haenisch B, Bönisch H, Cichon S, Allam JP, Novak N, Molderings GJEffects of exogenous agmatine in human leukemia HMC-1 and HL-60 cells on proliferation, polyamine metabolism and cell cycleLeuk Res.(2011 Sep)
36.^Regunathan S, Youngson C, Raasch W, Wang H, Reis DJImidazoline receptors and agmatine in blood vessels: a novel system inhibiting vascular smooth muscle proliferationJ Pharmacol Exp Ther.(1996 Mar)
39.^Otake K, Ruggiero DA, Regunathan S, Wang H, Milner TA, Reis DJRegional localization of agmatine in the rat brain: an immunocytochemical studyBrain Res.(1998 Mar 16)
41.^Wang H, Regunathan S, Youngson C, Bramwell S, Reis DJAn antibody to agmatine localizes the amine in bovine adrenal chromaffin cellsNeurosci Lett.(1995 Jan 2)
42.^Molderings GJ, Heinen A, Menzel S, Lübbecke F, Homann J, Göthert MGastrointestinal uptake of agmatine: distribution in tissues and organs and pathophysiologic relevanceAnn N Y Acad Sci.(2003 Dec)
43.^Gilad GM, Wollam Y, Iaina A, Rabey JM, Chernihovsky T, Gilad VHMetabolism of agmatine into urea but not into nitric oxide in rat brainNeuroreport.(1996 Jul 29)
45.^Yokoi I, Kabuto H, Habu H, Inada K, Toma J, Mori AStructure-activity relationships of arginine analogues on nitric oxide synthase activity in the rat brainNeuropharmacology.(1994 Nov)
47.^Novotny WF, Chassande O, Baker M, Lazdunski M, Barbry PDiamine oxidase is the amiloride-binding protein and is inhibited by amiloride analoguesJ Biol Chem.(1994 Apr 1)
48.^McGrath AP, Hilmer KM, Collyer CA, Dooley DM, Guss JMA new crystal form of human diamine oxidaseActa Crystallogr Sect F Struct Biol Cryst Commun.(2010 Feb 1)
49.^Cabella C, Gardini G, Corpillo D, Testore G, Bedino S, Solinas SP, Cravanzola C, Vargiu C, Grillo MA, Colombatto STransport and metabolism of agmatine in rat hepatocyte culturesEur J Biochem.(2001 Feb)
50.^Mella C, Martínez F, de Los Angeles García M, Nualart F, Castro V, Bustos P, Carvajal N, Uribe EExpression and localization of an agmatinase-like protein in the rat brainHistochem Cell Biol.(2010 Aug)
52.^Satriano J, Matsufuji S, Murakami Y, Lortie MJ, Schwartz D, Kelly CJ, Hayashi S, Blantz RCAgmatine suppresses proliferation by frameshift induction of antizyme and attenuation of cellular polyamine levelsJ Biol Chem.(1998 Jun 19)
53.^Babál P, Ruchko M, Campbell CC, Gilmour SP, Mitchell JL, Olson JW, Gillespie MNRegulation of ornithine decarboxylase activity and polyamine transport by agmatine in rat pulmonary artery endothelial cellsJ Pharmacol Exp Ther.(2001 Feb)
54.^Satriano J, Kelly CJ, Blantz RCAn emerging role for agmatineKidney Int.(1999 Oct)
56.^Wargnies B, Lauwers N, Stalon VStructure and properties of the putrescine carbamoyltransferase of Streptococcus faecalisEur J Biochem.(1979 Nov 1)
62.^Aricioglu F, Regunathan S, Piletz JEIs agmatine an endogenous factor against stressAnn N Y Acad Sci.(2003 Dec)
63.^Piletz JE, May PJ, Wang G, Zhu HAgmatine crosses the blood-brain barrierAnn N Y Acad Sci.(2003 Dec)
65.^Regunathan S, Dozier D, Takkalapalli R, Phillips WJAgmatine levels in the cerebrospinal fluid of normal human volunteersJ Pain Palliat Care Pharmacother.(2009)
66.^Jo I, Han C, Ahn Jo S, Seo JA, Park MH, Kim NHLow levels of plasma agmatine in the metabolic syndromeMetab Syndr Relat Disord.(2010 Feb)
67.^Li G, Regunathan S, Barrow CJ, Eshraghi J, Cooper R, Reis DJAgmatine: an endogenous clonidine-displacing substance in the brainScience.(1994 Feb 18)
68.^Uzbay T, Goktalay G, Kayir H, Eker SS, Sarandol A, Oral S, Buyukuysal L, Ulusoy G, Kirli SIncreased plasma agmatine levels in patients with schizophreniaJ Psychiatr Res.(2013 May 7)
69.^Halaris A, Zhu H, Feng Y, Piletz JEPlasma agmatine and platelet imidazoline receptors in depressionAnn N Y Acad Sci.(1999 Jun 21)
71.^Li YF, Gong ZH, Cao JB, Wang HL, Luo ZP, Li JAntidepressant-like effect of agmatine and its possible mechanismEur J Pharmacol.(2003 May 23)
72.^Roberts JC, Grocholski BM, Kitto KF, Fairbanks CAPharmacodynamic and pharmacokinetic studies of agmatine after spinal administration in the mouseJ Pharmacol Exp Ther.(2005 Sep)
74.^Satriano J, Isome M, Casero RA Jr, Thomson SC, Blantz RCPolyamine transport system mediates agmatine transport in mammalian cellsAm J Physiol Cell Physiol.(2001 Jul)
78.^Reis DJ, Regunathan SAgmatine: a novel neurotransmitterAdv Pharmacol.(1998)
79.^Sastre M, Regunathan S, Galea E, Reis DJAgmatinase activity in rat brain: a metabolic pathway for the degradation of agmatineJ Neurochem.(1996 Oct)
80.^Goracke-Postle CJ, Nguyen HO, Stone LS, Fairbanks CARelease of tritiated agmatine from spinal synaptosomesNeuroreport.(2006 Jan 23)
81.^Sastre M, Regunathan S, Reis DJUptake of agmatine into rat brain synaptosomes: possible role of cation channelsJ Neurochem.(1997 Dec)
82.^Goracke-Postle CJ, Overland AC, Riedl MS, Stone LS, Fairbanks CAPotassium- and capsaicin-induced release of agmatine from spinal nerve terminalsJ Neurochem.(2007 Sep)
83.^Goracke-Postle CJ, Overland AC, Stone LS, Fairbanks CAAgmatine transport into spinal nerve terminals is modulated by polyamine analogsJ Neurochem.(2007 Jan)
84.^Regunathan S, Feinstein DL, Raasch W, Reis DJAgmatine (decarboxylated arginine) is synthesized and stored in astrocytesNeuroreport.(1995 Oct 2)
85.^Zomkowski AD, Santos AR, Rodrigues ALPutrescine produces antidepressant-like effects in the forced swimming test and in the tail suspension test in miceProg Neuropsychopharmacol Biol Psychiatry.(2006 Dec 30)
86.^Molderings GJ, Menzel S, Kathmann M, Schlicker E, Göthert MDual interaction of agmatine with the rat alpha(2D)-adrenoceptor: competitive antagonism and allosteric activationBr J Pharmacol.(2000 Aug)
87.^Dias Elpo Zomkowski A, Oscar Rosa A, Lin J, Santos AR, Calixto JB, Lúcia Severo Rodrigues AEvidence for serotonin receptor subtypes involvement in agmatine antidepressant like-effect in the mouse forced swimming testBrain Res.(2004 Oct 15)
89.^Lewin AH, Sun G, Fudala L, Navarro H, Zhou LM, Popik P, Faynsteyn A, Skolnick PMolecular features associated with polyamine modulation of NMDA receptorsJ Med Chem.(1998 Mar 12)
92.^Morrissey JJ, Klahr SAgmatine activation of nitric oxide synthase in endothelial cellsProc Assoc Am Physicians.(1997 Jan)
95.^Pinthong D, Wright IK, Hanmer C, Millns P, Mason R, Kendall DA, Wilson VGAgmatine recognizes alpha 2-adrenoceptor binding sites but neither activates nor inhibits alpha 2-adrenoceptorsNaunyn Schmiedebergs Arch Pharmacol.(1995 Jan)
97.^Taksande BG, Kotagale NR, Nakhate KT, Mali PD, Kokare DM, Hirani K, Subhedar NK, Chopde CT, Ugale RRAgmatine in the hypothalamic paraventricular nucleus stimulates feeding in rats: involvement of neuropeptide YBr J Pharmacol.(2011 Sep)
98.^Zomkowski AD, Hammes L, Lin J, Calixto JB, Santos AR, Rodrigues ALAgmatine produces antidepressant-like effects in two models of depression in miceNeuroreport.(2002 Mar 25)
100.^Raasch W, Muhle H, Dominiak PModulation of MAO activity by imidazoline and guanidine derivativesAnn N Y Acad Sci.(1999 Jun 21)
102.^Head GA, Mayorov DNImidazoline receptors, novel agents and therapeutic potentialCardiovasc Hematol Agents Med Chem.(2006 Jan)
103.^González C, Regunathan S, Reis DJ, Estrada CAgmatine, an endogenous modulator of noradrenergic neurotransmission in the rat tail arteryBr J Pharmacol.(1996 Oct)
105.^Askalany AR, Yamakura T, Petrenko AB, Kohno T, Sakimura K, Baba HEffect of agmatine on heteromeric N-methyl-D-aspartate receptor channelsNeurosci Res.(2005 Aug)
108.^Anis N, Sherby S, Goodnow R Jr, Niwa M, Konno K, Kallimopoulos T, Bukownik R, Nakanishi K, Usherwood P, Eldefrawi A, et alStructure-activity relationships of philanthotoxin analogs and polyamines on N-methyl-D-aspartate and nicotinic acetylcholine receptorsJ Pharmacol Exp Ther.(1990 Sep)
109.^Zhu MY, Piletz JE, Halaris A, Regunathan SEffect of agmatine against cell death induced by NMDA and glutamate in neurons and PC12 cellsCell Mol Neurobiol.(2003 Oct)
110.^Brown RE, Stevens DR, Haas HLThe physiology of brain histamineProg Neurobiol.(2001 Apr)
111.^Burban A, Faucard R, Armand V, Bayard C, Vorobjev V, Arrang JMHistamine potentiates N-methyl-D-aspartate receptors by interacting with an allosteric site distinct from the polyamine binding siteJ Pharmacol Exp Ther.(2010 Mar)
112.^Watanabe C, Orito T, Watanabe H, Mizoguchi H, Yonezawa A, Yanai K, Mobarakeh JI, Onodera K, Sakurada T, Sakurada SIntrathecal high-dose histamine induces spinally-mediated nociceptive behavioral responses through a polyamine site of NMDA receptorsEur J Pharmacol.(2008 Feb 26)
113.^Wade CL, Eskridge LL, Nguyen HO, Kitto KF, Stone LS, Wilcox G, Fairbanks CAImmunoneutralization of agmatine sensitizes mice to micro-opioid receptor toleranceJ Pharmacol Exp Ther.(2009 Nov)
114.^Lewis B, Wellmann KA, Barron SAgmatine reduces balance deficits in a rat model of third trimester binge-like ethanol exposurePharmacol Biochem Behav.(2007 Nov)
115.^Barron S, Lewis B, Wellmann K, Carter M, Farook J, Ring J, Rogers DT, Holley R, Crooks P, Littleton JPolyamine modulation of NMDARs as a mechanism to reduce effects of alcohol dependenceRecent Pat CNS Drug Discov.(2012 Aug)
117.^Demady DR, Jianmongkol S, Vuletich JL, Bender AT, Osawa YAgmatine enhances the NADPH oxidase activity of neuronal NO synthase and leads to oxidative inactivation of the enzymeMol Pharmacol.(2001 Jan)
122.^Kolesnikov YA, Pick CG, Ciszewska G, Pasternak GWBlockade of tolerance to morphine but not to kappa opioids by a nitric oxide synthase inhibitorProc Natl Acad Sci U S A.(1993 Jun 1)
124.^Elliott K, Kest B, Man A, Kao B, Inturrisi CEN-methyl-D-aspartate (NMDA) receptors, mu and kappa opioid tolerance, and perspectives on new analgesic drug developmentNeuropsychopharmacology.(1995 Dec)
126.^Krass M, Wegener G, Vasar E, Volke VAntidepressant-like effect of agmatine is not mediated by serotoninBehav Brain Res.(2008 Apr 9)
127.^Molderings GJ, Schmidt K, Bönisch H, Göthert MInhibition of 5-HT3 receptor function by imidazolines in mouse neuroblastoma cells: potential involvement of sigma 2 binding sitesNaunyn Schmiedebergs Arch Pharmacol.(1996 Aug-Sep)
128.^Taksande BG, Kotagale NR, Tripathi SJ, Ugale RR, Chopde CTAntidepressant like effect of selective serotonin reuptake inhibitors involve modulation of imidazoline receptors by agmatineNeuropharmacology.(2009 Sep)
131.^Kotagale NR, Tripathi SJ, Aglawe MM, Chopde CT, Umekar MJ, Taksande BGEvidences for the agmatine involvement in antidepressant like effect of bupropion in mouse forced swim testPharmacol Biochem Behav.(2013 Jun)
132.^Tsou K, Brown S, Sañudo-Peña MC, Mackie K, Walker JMImmunohistochemical distribution of cannabinoid CB1 receptors in the rat central nervous systemNeuroscience.(1998 Mar)
133.^Pettit DA, Harrison MP, Olson JM, Spencer RF, Cabral GAImmunohistochemical localization of the neural cannabinoid receptor in rat brainJ Neurosci Res.(1998 Feb 1)
134.^Ruggiero DA, Regunathan S, Wang H, Milner TA, Reis DJImmunocytochemical localization of an imidazoline receptor protein in the central nervous systemBrain Res.(1998 Jan 12)
135.^Molderings GJ, Likungu J, Göthert MPresynaptic cannabinoid and imidazoline receptors in the human heart and their potential relationshipNaunyn Schmiedebergs Arch Pharmacol.(1999 Aug)
136.^Aggarwal S, Shavalian B, Kim E, Rawls SMAgmatine enhances cannabinoid action in the hot-plate assay of thermal nociceptionPharmacol Biochem Behav.(2009 Oct)
137.^Rawls SM, Tallarida RJ, Zisk JAgmatine and a cannabinoid agonist, WIN 55212-2, interact to produce a hypothermic synergyEur J Pharmacol.(2006 Dec 28)
140.^Spina E, Trovati A, Parolaro D, Giagnoni GA role of nitric oxide in WIN 55,212-2 tolerance in miceEur J Pharmacol.(1998 Feb 19)
141.^Molderings GJ, Likungu J, Jakschik J, Göthert MPresynaptic imidazoline receptors and non-adrenoceptor {3H}-idazoxan binding sites in human cardiovascular tissuesBr J Pharmacol.(1997 Sep)
143.^Li J, Li X, Pei G, Qin BYEffects of agmatine on tolerance to and substance dependence on morphine in miceZhongguo Yao Li Xue Bao.(1999 Mar)
144.^Kolesnikov Y, Jain S, Pasternak GWModulation of opioid analgesia by agmatineEur J Pharmacol.(1996 Jan 18)
147.^Su RB, Lu XQ, Huang Y, Liu Y, Gong ZH, Wei XL, Wu N, Li JEffects of intragastric agmatine on morphine-induced physiological dependence in beagle dogs and rhesus monkeysEur J Pharmacol.(2008 Jun 10)
148.^Gold MS, Redmond DE Jr, Kleber HDClonidine blocks acute opiate-withdrawal symptomsLancet.(1978 Sep 16)
151.^Iglesias V, Alguacil LF, Alamo C, Cuenca EEffects of yohimbine on morphine analgesia and physical dependence in the ratEur J Pharmacol.(1992 Jan 28)
152.^Dwoskin LP, Neal BS, Sparber SBYohimbine exacerbates and clonidine attenuates acute morphine withdrawal in ratsEur J Pharmacol.(1983 Jun 3)
153.^Wade CL, Schuster DJ, Domingo KM, Kitto KF, Fairbanks CASupraspinally-administered agmatine attenuates the development of oral fentanyl self-administrationEur J Pharmacol.(2008 Jun 10)
154.^Morgan AD, Campbell UC, Fons RD, Carroll MEEffects of agmatine on the escalation of intravenous cocaine and fentanyl self-administration in ratsPharmacol Biochem Behav.(2002 Jul)
155.^Huston JP, Silva MA, Topic B, Müller CPWhat's conditioned in conditioned place preferenceTrends Pharmacol Sci.(2013 Mar)
156.^Tahsili-Fahadan P, Yahyavi-Firouz-Abadi N, Khoshnoodi MA, Motiei-Langroudi R, Tahaei SA, Ghahremani MH, Dehpour ARAgmatine potentiates morphine-induced conditioned place preference in mice: modulation by alpha2-adrenoceptorsNeuropsychopharmacology.(2006 Aug)
157.^Yu Y, Chen Z, Li WG, Cao H, Feng EG, Yu F, Liu H, Jiang H, Xu TLA nonproton ligand sensor in the acid-sensing ion channelNeuron.(2010 Oct 6)
159.^Wemmie JA, Price MP, Welsh MJAcid-sensing ion channels: advances, questions and therapeutic opportunitiesTrends Neurosci.(2006 Oct)
160.^Waldmann R, Champigny G, Bassilana F, Heurteaux C, Lazdunski MA proton-gated cation channel involved in acid-sensingNature.(1997 Mar 13)
162.^Salinas M, Lazdunski M, Lingueglia EStructural elements for the generation of sustained currents by the acid pain sensor ASIC3J Biol Chem.(2009 Nov 13)
163.^Deval E, Noël J, Lay N, Alloui A, Diochot S, Friend V, Jodar M, Lazdunski M, Lingueglia EASIC3, a sensor of acidic and primary inflammatory painEMBO J.(2008 Nov 19)
164.^Li WG, Xu TLASIC3 channels in multimodal sensory perceptionACS Chem Neurosci.(2011 Jan 19)
166.^Fairbanks CA, Schreiber KL, Brewer KL, Yu CG, Stone LS, Kitto KF, Nguyen HO, Grocholski BM, Shoeman DW, Kehl LJ, Regunathan S, Reis DJ, Yezierski RP, Wilcox GLAgmatine reverses pain induced by inflammation, neuropathy, and spinal cord injuryProc Natl Acad Sci U S A.(2000 Sep 12)
167.^Su RB, Li J, Qin BYA biphasic opioid function modulator: agmatineActa Pharmacol Sin.(2003 Jul)
170.^Gourlay S, Forbes A, Marriner T, Kutin J, McNeil JA placebo-controlled study of three clonidine doses for smoking cessationClin Pharmacol Ther.(1994 Jan)
172.^Kotagale NR, Taksande BG, Gahane AY, Ugale RR, Chopde CTRepeated agmatine treatment attenuates nicotine sensitization in mice: modulation by alpha2-adrenoceptorsBehav Brain Res.(2010 Dec 1)
173.^Zaniewska M, McCreary AC, Sezer G, Przegaliński E, Filip MEffects of agmatine on nicotine-evoked behavioral responses in ratsPharmacol Rep.(2008 Sep-Oct)
174.^Uzbay IT, Yeşilyurt O, Celik T, Ergün H, Işimer AEffects of agmatine on ethanol withdrawal syndrome in ratsBehav Brain Res.(2000 Jan)
175.^Taksande BG, Kotagale NR, Patel MR, Shelkar GP, Ugale RR, Chopde CTAgmatine, an endogenous imidazoline receptor ligand modulates ethanol anxiolysis and withdrawal anxiety in ratsEur J Pharmacol.(2010 Jul 10)
176.^Littleton JM, Lovinger D, Liljequist S, Ticku R, Matsumoto I, Barron SRole of polyamines and NMDA receptors in ethanol dependence and withdrawalAlcohol Clin Exp Res.(2001 May)
177.^Gibson DA, Harris BR, Prendergast MA, Hart SR, Blanchard JA 2nd, Holley RC, Pedigo NW, Littleton JMPolyamines contribute to ethanol withdrawal-induced neurotoxicity in rat hippocampal slice cultures through interactions with the NMDA receptorAlcohol Clin Exp Res.(2003 Jul)
178.^Collins ED, Ward AS, McDowell DM, Foltin RW, Fischman MWThe effects of memantine on the subjective, reinforcing and cardiovascular effects of cocaine in humansBehav Pharmacol.(1998 Nov)
179.^Collins ED, Vosberg SK, Ward AS, Haney M, Foltin RWThe effects of acute pretreatment with high-dose memantine on the cardiovascular and behavioral effects of cocaine in humansExp Clin Psychopharmacol.(2007 Jun)
181.^Cantin L, Lenoir M, Augier E, Vanhille N, Dubreucq S, Serre F, Vouillac C, Ahmed SHCocaine is low on the value ladder of rats: possible evidence for resilience to addictionPLoS One.(2010 Jul 28)
182.^Lenoir M, Serre F, Cantin L, Ahmed SHIntense sweetness surpasses cocaine rewardPLoS One.(2007 Aug 1)
185.^Kim JH, Lee YW, Park KA, Lee WT, Lee JEAgmatine attenuates brain edema through reducing the expression of aquaporin-1 after cerebral ischemiaJ Cereb Blood Flow Metab.(2010 May)
190.^Moro MA, Cárdenas A, Hurtado O, Leza JC, Lizasoain IRole of nitric oxide after brain ischaemiaCell Calcium.(2004 Sep-Oct)
192.^Huang Z, Huang PL, Panahian N, Dalkara T, Fishman MC, Moskowitz MAEffects of cerebral ischemia in mice deficient in neuronal nitric oxide synthaseScience.(1994 Sep 23)
194.^Ma T, Yang B, Gillespie A, Carlson EJ, Epstein CJ, Verkman ASGeneration and phenotype of a transgenic knockout mouse lacking the mercurial-insensitive water channel aquaporin-4J Clin Invest.(1997 Sep 1)
195.^Manley GT, Fujimura M, Ma T, Noshita N, Filiz F, Bollen AW, Chan P, Verkman ASAquaporin-4 deletion in mice reduces brain edema after acute water intoxication and ischemic strokeNat Med.(2000 Feb)
196.^Ribeiro Mde C, Hirt L, Bogousslavsky J, Regli L, Badaut JTime course of aquaporin expression after transient focal cerebral ischemia in miceJ Neurosci Res.(2006 May 15)
197.^Gilad GM, Salame K, Rabey JM, Gilad VHAgmatine treatment is neuroprotective in rodent brain injury modelsLife Sci.(1996)
202.^Kanwar JR, Kamalapuram SK, Kanwar RKTargeting survivin in cancer: the cell-signalling perspectiveDrug Discov Today.(2011 Jun)
203.^Lin J, Guan Z, Wang C, Feng L, Zheng Y, Caicedo E, Bearth E, Peng JR, Gaffney P, Ondrey FGInhibitor of differentiation 1 contributes to head and neck squamous cell carcinoma survival via the NF-kappaB/survivin and phosphoinositide 3-kinase/Akt signaling pathwaysClin Cancer Res.(2010 Jan 1)
207.^Xu X, Shi G, Jia RChanges of polyamine levels in roots of Sagittaria sagittifolia L. under copper stressEnviron Sci Pollut Res Int.(2011 Aug)
208.^Ruiz-Durántez E, Ruiz-Ortega JA, Pineda J, Ugedo LEffect of agmatine on locus coeruleus neuron activity: possible involvement of nitric oxideBr J Pharmacol.(2002 Mar)
209.^Weiss JM, Stout JC, Aaron MF, Quan N, Owens MJ, Butler PD, Nemeroff CBDepression and anxiety: role of the locus coeruleus and corticotropin-releasing factorBrain Res Bull.(1994)
210.^Lavinsky D, Arteni NS, Netto CAAgmatine induces anxiolysis in the elevated plus maze task in adult ratsBehav Brain Res.(2003 Apr 17)
211.^Aricioglu F, Altunbas HIs agmatine an endogenous anxiolytic/antidepressant agentAnn N Y Acad Sci.(2003 Dec)
212.^Gong ZH, Li YF, Zhao N, Yang HJ, Su RB, Luo ZP, Li JAnxiolytic effect of agmatine in rats and miceEur J Pharmacol.(2006 Nov 21)
216.^Budni J, Gadotti VM, Kaster MP, Santos AR, Rodrigues ALRole of different types of potassium channels in the antidepressant-like effect of agmatine in the mouse forced swimming testEur J Pharmacol.(2007 Dec 1)
220.^Kobayashi T, Washiyama K, Ikeda KInhibition of G protein-activated inwardly rectifying K+ channels by various antidepressant drugsNeuropsychopharmacology.(2004 Oct)
221.^Kaster MP, Budni J, Binfaré RW, Santos AR, Rodrigues ALThe inhibition of different types of potassium channels underlies the antidepressant-like effect of adenosine in the mouse forced swimming testProg Neuropsychopharmacol Biol Psychiatry.(2007 Apr 13)
222.^Budni J, Freitas AE, Binfaré RW, Rodrigues ALRole of potassium channels in the antidepressant-like effect of folic acid in the forced swimming test in micePharmacol Biochem Behav.(2012 Mar)
224.^Bortolatto CF, Jesse CR, Wilhelm EA, Nogueira CWInvolvement of potassium channels in the antidepressant-like effect of venlafaxine in miceLife Sci.(2010 Feb 27)
225.^Kelly JS, Larkman P, Penington NJ, Rainnie DG, McAllister-Williams H, Hodgkiss JSerotonin receptor heterogeneity and the role of potassium channels in neuronal excitabilityAdv Exp Med Biol.(1991)
226.^Weng XC, Gai XD, Zheng JQ, Li JAgmatine blocked voltage-gated calcium channel in cultured rat hippocampal neuronsActa Pharmacol Sin.(2003 Aug)
228.^Pedrazzini T, Pralong F, Grouzmann ENeuropeptide Y: the universal soldierCell Mol Life Sci.(2003 Feb)
232.^Everitt BJ, Hökfelt T, Terenius L, Tatemoto K, Mutt V, Goldstein MDifferential co-existence of neuropeptide Y (NPY)-like immunoreactivity with catecholamines in the central nervous system of the ratNeuroscience.(1984 Feb)
234.^Smiałowska M, Obuchowicz E, Turchan J, Herman ZS, Przewłocki RClonidine administration increases neuropeptide Y immunoreactivity and neuropeptide Y mRNA in the rat cerebral cortex neuronsNeuropeptides.(1997 Jun)
239.^Betancourt L, Rada P, Paredes D, Hernández LIn vivo monitoring of cerebral agmatine by microdialysis and capillary electrophoresisJ Chromatogr B Analyt Technol Biomed Life Sci.(2012 Jan 1)
240.^Liu P, Collie ND, Chary S, Jing Y, Zhang HSpatial learning results in elevated agmatine levels in the rat brainHippocampus.(2008)
241.^Leitch B, Shevtsova O, Reusch K, Bergin DH, Liu PSpatial learning-induced increase in agmatine levels at hippocampal CA1 synapsesSynapse.(2011 Feb)
242.^Rushaidhi M, Jing Y, Zhang H, Liu PParticipation of hippocampal agmatine in spatial learning: an in vivo microdialysis studyNeuropharmacology.(2013 Feb)
243.^Wang ZM, Sun GQ, Wang ZA, He RREffects of agmatine on neuronal discharges in rat hippocampal CA1 areaSheng Li Xue Bao.(2003 Dec 25)
244.^Liu P, Jing Y, Collie ND, Chary S, Zhang HMemory-related changes in L-citrulline and agmatine in the rat brainHippocampus.(2009 Jul)
245.^Arteni NS, Lavinsky D, Rodrigues AL, Frison VB, Netto CAAgmatine facilitates memory of an inhibitory avoidance task in adult ratsNeurobiol Learn Mem.(2002 Sep)
247.^Dalley JW, Cardinal RN, Robbins TWPrefrontal executive and cognitive functions in rodents: neural and neurochemical substratesNeurosci Biobehav Rev.(2004 Nov)
249.^Riccio A, Ahn S, Davenport CM, Blendy JA, Ginty DDMediation by a CREB family transcription factor of NGF-dependent survival of sympathetic neuronsScience.(1999 Dec 17)
250.^Du K, Montminy MCREB is a regulatory target for the protein kinase Akt/PKBJ Biol Chem.(1998 Dec 4)
251.^Chen X, Li Y, Wang L, Katakowski M, Zhang L, Chen J, Xu Y, Gautam SC, Chopp MIschemic rat brain extracts induce human marrow stromal cell growth factor productionNeuropathology.(2002 Dec)
252.^Lu Y, Christian K, Lu BBDNF: a key regulator for protein synthesis-dependent LTP and long-term memoryNeurobiol Learn Mem.(2008 Mar)
253.^Collingridge GL, Bliss TVMemories of NMDA receptors and LTPTrends Neurosci.(1995 Feb)
254.^Cain DPLTP, NMDA, genes and learningCurr Opin Neurobiol.(1997 Apr)
257.^Rastegar K, Roosta H, Zarifkar A, Rafati A, Moosavi MThe Effect of Intra-CA1 Agmatine Microinjection on Water Maze Learning and Memory in RatIran Red Crescent Med J.(2011 May)
259.^McKay BE, Lado WE, Martin LJ, Galic MA, Fournier NMLearning and memory in agmatine-treated ratsPharmacol Biochem Behav.(2002 Jun)
260.^Rushaidhi M, Collie ND, Zhang H, Liu PAgmatine selectively improves behavioural function in aged male Sprague-Dawley ratsNeuroscience.(2012 Aug 30)
261.^Liu P, Rushaidhi M, Collie ND, Leong MT, Zhang HBehavioral effects of intracerebroventricular microinfusion of agmatine in adult ratsBehav Neurosci.(2008 Jun)
264.^Demehri S, Homayoun H, Honar H, Riazi K, Vafaie K, Roushanzamir F, Dehpour ARAgmatine exerts anticonvulsant effect in mice: modulation by alpha 2-adrenoceptors and nitric oxideNeuropharmacology.(2003 Sep)
265.^Bence AK, Worthen DR, Stables JP, Crooks PAAn in vivo evaluation of the antiseizure activity and acute neurotoxicity of agmatinePharmacol Biochem Behav.(2003 Feb)
266.^Luszczki JJ, Czernecki R, Wojtal K, Borowicz KK, Czuczwar SJAgmatine enhances the anticonvulsant action of phenobarbital and valproate in the mouse maximal electroshock seizure modelJ Neural Transm.(2008 Nov)
271.^Jeong SY, Ha TS, Park CS, Uhm DY, Chung SNitric oxide directly activates large conductance Ca2+-activated K+ channels (rSlo)Mol Cells.(2001 Aug 31)
273.^Denninger JW, Marletta MAGuanylate cyclase and the .NO/cGMP signaling pathwayBiochim Biophys Acta.(1999 May 5)
274.^Bhutada P, Mundhada Y, Humane V, Rahigude A, Deshmukh P, Latad S, Jain KAgmatine, an endogenous ligand of imidazoline receptor protects against memory impairment and biochemical alterations in streptozotocin-induced diabetic ratsProg Neuropsychopharmacol Biol Psychiatry.(2012 Apr 27)
277.^Matheus FC, Aguiar AS Jr, Castro AA, Villarinho JG, Ferreira J, Figueiredo CP, Walz R, Santos AR, Tasca CI, Prediger RDNeuroprotective effects of agmatine in mice infused with a single intranasal administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)Behav Brain Res.(2012 Dec 1)
278.^Moosavi M, Khales GY, Abbasi L, Zarifkar A, Rastegar KAgmatine protects against scopolamine-induced water maze performance impairment and hippocampal ERK and Akt inactivationNeuropharmacology.(2012 Apr)
279.^Utkan T, Gocmez SS, Regunathan S, Aricioglu FAgmatine, a metabolite of L-arginine, reverses scopolamine-induced learning and memory impairment in ratsPharmacol Biochem Behav.(2012 Oct)
280.^Zarifkar A, Choopani S, Ghasemi R, Naghdi N, Maghsoudi AH, Maghsoudi N, Rastegar K, Moosavi MAgmatine prevents LPS-induced spatial memory impairment and hippocampal apoptosisEur J Pharmacol.(2010 May 25)
281.^Ahn SK, Hong S, Park YM, Choi JY, Lee WT, Park KA, Lee JEProtective effects of agmatine on lipopolysaccharide-injured microglia and inducible nitric oxide synthase activityLife Sci.(2012 Dec 17)
282.^Kohl S, Heekeren K, Klosterkötter J, Kuhn JPrepulse inhibition in psychiatric disorders--apart from schizophreniaJ Psychiatr Res.(2013 Apr)
283.^Geyer MA, Krebs-Thomson K, Braff DL, Swerdlow NRPharmacological studies of prepulse inhibition models of sensorimotor gating deficits in schizophrenia: a decade in reviewPsychopharmacology (Berl).(2001 Jul)
285.^Uzbay T, Kayir H, Goktalay G, Yildirim MAgmatine disrupts prepulse inhibition of acoustic startle reflex in ratsJ Psychopharmacol.(2010 Jun)
286.^Pålsson E, Fejgin K, Wass C, Klamer DAgmatine attenuates the disruptive effects of phencyclidine on prepulse inhibitionEur J Pharmacol.(2008 Aug 20)
287.^Kotagale NR, Taksande BG, Wadhwani PJ, Palhade MW, Mendhi SM, Gawande DY, Hadole PN, Chopde CTPsychopharmacological study of agmatine in behavioral tests of schizophrenia in rodentsPharmacol Biochem Behav.(2012 Jan)
288.^Liu P, Smith PF, Appleton I, Darlington CL, Bilkey DKAge-related changes in nitric oxide synthase and arginase in the rat prefrontal cortexNeurobiol Aging.(2004 Apr)
289.^Liu P, Chary S, Devaraj R, Jing Y, Darlington CL, Smith PF, Tucker IG, Zhang HEffects of aging on agmatine levels in memory-associated brain structuresHippocampus.(2008)
290.^Liu P, Gupta N, Jing Y, Zhang HAge-related changes in polyamines in memory-associated brain structures in ratsNeuroscience.(2008 Aug 26)
291.^Gupta N, Jing Y, Collie ND, Zhang H, Liu PAgeing alters behavioural function and brain arginine metabolism in male Sprague-Dawley ratsNeuroscience.(2012 Dec 13)
292.^Liu P, Smith PF, Appleton I, Darlington CL, Bilkey DKPotential involvement of NOS and arginase in age-related behavioural impairmentsExp Gerontol.(2004 Aug)
293.^Liu P, Smith PF, Appleton I, Darlington CL, Bilkey DKHippocampal nitric oxide synthase and arginase and age-associated behavioral deficitsHippocampus.(2005)
294.^Rushaidhi M, Zhang H, Liu PEffects of prolonged agmatine treatment in aged male Sprague-Dawley ratsNeuroscience.(2013 Mar 27)
295.^Joshi MS, Ferguson TB Jr, Johnson FK, Johnson RA, Parthasarathy S, Lancaster JR JrReceptor-mediated activation of nitric oxide synthesis by arginine in endothelial cellsProc Natl Acad Sci U S A.(2007 Jun 12)
296.^Haulică I, Bild W, Iliescu R, Georgescu R, Frunză FPreliminary research on possible relationship of NO with agmatine at the vascular levelRom J Physiol.(1999 Jan-Jun)
297.^Schwartz D, Peterson OW, Mendonca M, Satriano J, Lortie M, Blantz RCAgmatine affects glomerular filtration via a nitric oxide synthase-dependent mechanismAm J Physiol.(1997 May)
298.^Fill M, Copello JARyanodine receptor calcium release channelsPhysiol Rev.(2002 Oct)
300.^Stoyanovsky D, Murphy T, Anno PR, Kim YM, Salama GNitric oxide activates skeletal and cardiac ryanodine receptorsCell Calcium.(1997 Jan)
302.^Lee HC, Galione A, Walseth TFCyclic ADP-ribose: metabolism and calcium mobilizing functionVitam Horm.(1994)
303.^Satriano J, Cunard R, Peterson OW, Dousa T, Gabbai FB, Blantz RCEffects on kidney filtration rate by agmatine requires activation of ryanodine channels for nitric oxide generationAm J Physiol Renal Physiol.(2008 Apr)
304.^Jagnandan D, Sessa WC, Fulton DIntracellular location regulates calcium-calmodulin-dependent activation of organelle-restricted eNOSAm J Physiol Cell Physiol.(2005 Oct)
305.^Liou SF, Wu JR, Lai WT, Sheu SH, Chen IJ, Yeh JLThe vasorelaxing action of labedipinedilol-A involves endothelial cell-derived NO and eNOS expression caused by calcium influxJ Cardiovasc Pharmacol.(2005 Mar)
308.^Chen CT, Chen W, Chung HH, Cheng KC, Yeh CH, Cheng JTActivation of imidazoline I-2B receptor by metformin to increase glucose uptake in skeletal muscleHorm Metab Res.(2011 Sep)
310.^Lu XL, Xu ZL, Yao XL, Su FJ, Ye CH, Li J, Lin YC, Wang GL, Zeng JS, Huang RX, Ou JS, Sun HS, Wang LP, Pang JY, Pei ZMarine cyclotripeptide X-13 promotes angiogenesis in zebrafish and human endothelial cells via PI3K/Akt/eNOS signaling pathwaysMar Drugs.(2012 Jun)
312.^Jung HJ, Jeon YH, Bokara KK, Koo BN, Lee WT, Park KA, Lee JEAgmatine promotes the migration of murine brain endothelial cells via multiple signaling pathwaysLife Sci.(2013 Jan 17)
313.^Auguet M, Viossat I, Marin JG, Chabrier PESelective inhibition of inducible nitric oxide synthase by agmatineJpn J Pharmacol.(1995 Nov)
314.^Jianmongkol S, Vuletich JL, Bender AT, Demady DR, Osawa YAminoguanidine-mediated inactivation and alteration of neuronal nitric-oxide synthaseJ Biol Chem.(2000 May 5)
316.^Nakatsuka M, Nakatsuka K, Osawa YMetabolism-based inactivation of penile nitric oxide synthase activity by guanabenzDrug Metab Dispos.(1998 May)
317.^Li Q, Shang ZL, Yin JX, Wang YH, He RREffect of agmatine on intracellular free calcium concentration in isolated rat ventricular myocytesSheng Li Xue Bao.(2002 Dec 25)
318.^Li Q, Yin JX, He RREffect of agmatine on L-type calcium current in rat ventricular myocytesActa Pharmacol Sin.(2002 Mar)
319.^Lortie MJ, Novotny WF, Peterson OW, Vallon V, Malvey K, Mendonca M, Satriano J, Insel P, Thomson SC, Blantz RCAgmatine, a bioactive metabolite of arginine. Production, degradation, and functional effects in the kidney of the ratJ Clin Invest.(1996 Jan 15)
320.^Gao Y, Gumusel B, Koves G, Prasad A, Hao Q, Hyman A, Lippton HAgmatine: a novel endogenous vasodilator substanceLife Sci.(1995)
321.^Ishikawa T, Misonou T, Ikeno M, Sato K, Sakamaki TN omega-hydroxyagmatine: a novel substance causing endothelium-dependent vasorelaxationBiochem Biophys Res Commun.(1995 Sep 5)
322.^Gill F, Pelit T, Terzioğlu B, Ekinci O, Gören MZEffects of agmatine on the survival rate in rats bled to hemorrhageArzneimittelforschung.(2011)
324.^Ernsberger P, Giuliano R, Willette RN, Reis DJRole of imidazole receptors in the vasodepressor response to clonidine analogs in the rostral ventrolateral medullaJ Pharmacol Exp Ther.(1990 Apr)
326.^Schäfer U, Raasch W, Qadri F, Chun J, Dominiak PEffects of agmatine on the cardiovascular system of spontaneously hypertensive ratsAnn N Y Acad Sci.(1999 Jun 21)
327.^Sun MK, Regunathan S, Reis DJCardiovascular responses to agmatine, a clonidine-displacing substance, in anesthetized ratClin Exp Hypertens.(1995 Jan-Feb)
329.^Cheng JT, Liu IM, Tzeng TF, Tsai CC, Lai TYPlasma glucose-lowering effect of beta-endorphin in streptozotocin-induced diabetic ratsHorm Metab Res.(2002 Oct)
331.^Radosevich PM, Williams PE, McRae JR, Lacy WW, Orth DN, Abumrad NNBeta-endorphin inhibits glucose production in the conscious dogJ Clin Invest.(1984 Apr)
332.^Curry DL, Bennett LL, Li CHStimulation of insulin secretion by beta-endorphins (1-27 & 1-31)Life Sci.(1987 May 25)
333.^Hauner H, Glatting G, Ditschuneit HH, Pfeiffer EFEndogenous opiates do not influence glucose and lipid metabolism in rat adipocytesExp Clin Endocrinol.(1988 Aug)
336.^Hong S, Kim CY, Lee WS, Shim J, Yeom HY, Seong GJOcular hypotensive effects of topically administered agmatine in a chronic ocular hypertensive rat modelExp Eye Res.(2010 Jan)
339.^Utkan T, Ulak G, Yildiran HG, Yardimoglu M, Gacar MNInvestigation on the mechanism involved in the effects of agmatine on ethanol-induced gastric mucosal injury in ratsLife Sci.(2000 Mar 24)
342.^Zádori ZS, Fehér A, Al-Khrasani M, Lackó E, Tóth VE, Brancati SB, Hein L, Mátyus P, Gyires KImidazoline versus alpha2-adrenoceptors in the control of gastric motility in miceEur J Pharmacol.(2013 Apr 5)
343.^Molderings GJ, Burian M, Homann J, Nilius M, Göthert MPotential relevance of agmatine as a virulence factor of Helicobacter pyloriDig Dis Sci.(1999 Dec)
345.^Jurkiewicz NH, Garcez do Carmo L, Hirata H, da Costa Santos W, Jurkiewicz AFunctional properties of agmatine in rat vas deferensEur J Pharmacol.(1996 Jul 4)
346.^Santos WC, Smaili SS, Jurkiewicz A, Piçarro I, Garcez-do-Carmo LDual effect of agmatine in the bisected rat vas deferensJ Pharm Pharmacol.(2003 Mar)
348.^Aglawe MM, Taksande BG, Kuldhariya SS, Chopde CT, Umekar MJ, Kotagale NRParticipation of central imidazoline binding sites in antinociceptive effect of ethanol and nicotine in ratsFundam Clin Pharmacol.(2013 Apr 11)
349.^Ozden O, Kayir H, Ozturk Y, Uzbay TAgmatine blocks ethanol-induced locomotor hyperactivity in male miceEur J Pharmacol.(2011 Apr 4)
351.^Uzbay T, Kose A, Kayir H, Ulusoy G, Celik TSex-related effects of agmatine on caffeine-induced locomotor activity in Swiss Webster miceEur J Pharmacol.(2010 Mar 25)