Abstract
Our understanding of the complex signaling neurophysiology of the central nervous system has facilitated the exploration of potential novel receptor-ligand system targets for disorders of this most complex organ. In recent years, many relatively neglected receptor-ligand systems have been re-evaluated with respect to their ability to potently modulate discrete tracts in the central nervous system. One such system is the tachykinin (previously neurokinin) system. The multiple heptahelical G protein-coupled receptors and neuropeptide ligands that comprise this system may be significantly involved in more central nervous systems actions than previously thought, including sleep disorders, amyotrophic lateral sclerosis, Alzheimer’s and Machado-Joseph disease. The development of our understanding of the role of the tachykinin receptor-ligand system in higher order central functions is likely to allow the creation of more specific and selective tachykinin-related neurotherapeutics.
Keywords: central neurological disorders, substance P, neurokinin A, neurokinin B, neurotherapeutics, receptor-ligand systems, tachykinin
INTRODUCTION
Tachykinin (TK) peptide neurotransmitters form a large functional group of signaling peptides in mammals, amphibia, mollusks, and invertebrates. This functional peptide group is typified, as the name implies, by possessing a potent and rapid muscular contractile action. These peptides have been demonstrated to control multiple and diverse physiological functions in these species. In each of the multiple species that employ these transmissive peptides, the actions of the ligand are mediated via activation of multiple and distinct isoforms of rhodopsin-like, heptahelical G protein-coupled receptors (GPCRs). Despite the ligands and receptors possessing intriguing and important functions in non-mammalian species, in this review we will concentrate on the potential usefulness of pharmacological manipulation of the TK receptor-ligand system for human disorders of a neurological origin. The goals of this review are to outline the roles that TKs play in various diseases and disorders of the central nervous system (CNS) and to present the data supporting the potential therapeutic avenues TKs provide.
MAMMALIAN TACHYKININ PEPTIDES
Mammalian tachykinins, previously referred to as neurokinins, consist of a family of three primary functional peptides: substance P (SP), neurokinin A (NKA), and neurokinin B (NKB), as well as four more recently classified peptides, i.e. neuropeptide K [1], neuropeptide γ [2] and hemokinin-1 [3]. As this review primarily concerns the therapeutic actions and relevance of tachykinins to neurophysiology, we will restrict our discussion to evidence implicating the three primary TKs, SP, NKA, and NKB, in pathophysiology. These were originally discovered in 1931 by von Euler and Gaddum [4] as an unknown potent stimulator of muscular contractile substance found in horse brain and intestine. SP was the first discovered TK and ignited a lengthy history of research of TKs [4]. SP was further studied and purified over the following 30 years with little success until the first molecular characterization of a TK peptide was uncovered in a structurally unidentified substance within the salivary gland of a Mediterranean octopus (Eledone moschata) [5]. The amino acid sequence of SP was only identified in 1971, forty years after its initial discovery [6]. In the following five years, two more peptides were purified and elucidated as TKs, after which a number of structurally or functionally related peptides were found throughout the CNS of various vertebrates and amphibians, including NKA and NKB. These peptides were discovered in the porcine spinal cord [7, 8] and all contained a common COOH-terminal pentapeptide sequence Phe-X-Gly-Leu-Met-NH2 [9]. In mammals the TK peptides are mainly expressed in neuronal tissue; therefore, in addition to their potent muscular actions, it appeared highly likely that TKs would possess important actions in the CNS. It is now well appreciated that the three primary mammalian TKs (SP, NKA, NKB) can all function as physiologically-relevant neurotransmitters/neuromodulators. The TK peptides are all amidated at their carboxyl terminus methionine. The amino acid sequence of SP, NKA, and NKB is identical in all mammals, but the sequence of hemokinin-1 appears to vary among mice, rats, and humans [3, 10].
With respect to their molecular origin, all the mammalian TK peptides are derived from three discrete gene products: preprotachykinin A (PPTA), preprotachykinin B (PPTB), or preprotachykinin C. SP, NKA, neuropeptide K, and neuropeptide γ are all encoded by PPTA. NKB is the only TK encoded from the PPTB gene, while hemokinin-1 is produced from the PPTC gene [3, 10]. As the expression of the PPTA/B genes is widespread, it is unsurprising that TKs are expressed in multiple organs (small and large intestine, lung, kidney, heart) as well as neuronal tissue. In tissues of neural origin, TKs are expressed primarily in the CNS, including the hypothalamus in mammals, although they are present in the peripheral nervous system (PNS) as well [11].
MAMMALIAN TACHYKININ RECEPTORS
The primary biological actions of the TKs are mediated through the activation of three distinct heptahelical GPCR isoforms. The receptor systems through which these ligands act are also well characterized [12–14]. In mammals, there are three primary TK GPCRs: TACR1, TACR2 and TACR3 receptors. These receptors share between 40–50% identity at the amino acid level [11]. Each receptor demonstrates a preferential affinity for one of the TK peptides, i.e., SP-TACR1, NKA-TACR2, NKB-TACR3. All three peptides, however, can act as full agonists at each of the three receptors, although their potency order mimics their affinity specificity, i.e., SP is the most potent at TACR1, NKA is the most potent at the TACR2, and NKB is the most potent at the TACR3 receptor. Each of the three TK GPCRs can be effectively coupled to multiple intracellular signaling systems (adenylyl cyclase, phospholipase C-beta, calcium-dependent potassium channel modulation, transient receptor potential channel modulation to exert their eventual cellular effects [13, 15–18]. As the majority of the ligand and receptor components of the TK system are highly enriched in the CNS, we will dedicate the majority of this review to investigating the roles of the TK system in complex neurological disorders.
TACHYKININS AND CENTRAL NERVOUS SYSTEM DISORDERS
I. AFFECTIVE DISORDERS
Affective disorders are characterized by a consistent, pervasive alteration in mood and affecting thoughts, emotions, and behaviors. Affective disorders include depression, anxiety, and bi-polar disorder. In recent years, there has been a plethora of data indicating the involvement of tachykinins, particularly SP, in affective disorders [19, 20]. A number of approaches has been utilized to study the role that SP plays in the etiology of human affective disorders, including the measurement of serum levels in depressed as well as anxious patients, infusion of SP into otherwise healthy patients, and measurement of SP levels in patients suffering from depression. Schedlowski and colleagues [21] as well as Weiss and colleagues [22] demonstrated that subjects with high anxiety generally displayed higher plasma levels of SP than their low anxiety counterparts. In a 2003 study examining the SP levels of patients suffering from depression, Bondy and colleagues found that patients diagnosed with acute depression had significantly and consistently higher SP levels than their control counterparts over a four-week period [23]. In addition to studies of plasma levels of TKs, cerebrospinal fluid (CSF) levels of SP have also been directly correlated with depression [24]. Interestingly, it has also been shown that there is a direct correlation of CSF SP levels with the presence of schizophrenic symptomology [24]. Reinforcing the argument for the involvement of SP in depression, numerous additional studies have also reported significantly elevated levels of CSF SP in patients with fibromyalgia syndrome [25–27], a disorder that is strongly associated with depression [28]. Interestingly, infusion of SP during sleep has been shown to significantly worsen mood upon wakening, as well as decrease the quality of sleep. Subjects given intravenous infusions of SP displayed an increase in REM latency, increased stage 1 sleep, and longer time awake, all objective measurements of decreased sleep quality [29]. It appears, therefore, that TK ligands are phenomenologically and functionally linked to the control of affective phenotypes in a complex manner.
II. AMYOTROPHIC LATERAL SCLEROSIS AND MOTOR NEURON DISEASE
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder characterized by the progressive loss of motor neuron function, primarily via neuronal oxidative mechanisms. Associations between ALS and TKs were first investigated by Gillberg and colleagues in 1982 [30]. In the spinal cord of ALS and control patients, some distinct perturbations in the numbers of SP-positive cells were noted. Matsuishi and colleagues demonstrated an important connection between TKs and ALS etiology [31]. When CSF SP levels were measured in ALS patients and controls, no significant differences were found; however, when the researchers investigated differences in SP levels within the ALS subjects, it was found that patients who presented with ALS for fewer than 2.5 years had significantly higher levels than patients who had been afflicted for greater than 2.5 years. It was hypothesized that in this context, SP may be acting as a neurotrophic factor that is elevated in order to compensate for the degeneration of the anterior horn cells that occurs in ALS. In addition to ALS, perturbations in the TK system have also been shown to play a role in related disorders, such as adult-onset motor neuron disease with basophilic inclusions (MND/BI). Significant reductions of SP-immunopositive cells have been demonstrated in the substantia nigra of MND/BI patients compared to control subjects [32].
III. EPILEPSY
Multiple forms of epilepsy exist, and they are differentiated upon the type and severity of presented symptomology. One of the most debilitating forms of epilepsy is status epilepticus (SE). In SE, seizures occur in the form of a series of protracted episodes that can last for prolonged periods of time and can thus be fatal if episodes merge with each other and immediate treatment is not available. Both SP, and NKB to a lesser extent, have been implicated as possible causal factors in the generation of SE [33]. When injected into the rat hippocampus, SP can significantly lower, in a dose-dependent manner, the initiation threshold for seizures induced by perforant path stimulation (PPS) [34]. It was subsequently demonstrated that spantide II, a substance P receptor antagonist, was able to both suppress electro-encephalogram (EEG) spike frequency as well as prevent the development of SE induced by the PPS protocol. Additionally, reinforcing an important role of TKs in SE, genetically-ablated PPTA(−/−) mice display significantly higher resistance to seizures induced by kainate injection. The PPTA(−/−) mice have been shown to experience seizures of shorter duration and lower intensity, as well as recover more rapidly from episodes. Wild-type mice given the same dose of kainate exhibited many neurons with acidophilic cytoplasm and shrunken, pyknotic nuclei in the hippocampus [35]. These data indicate a pro-convulsant role for the PPTA gene and thus SP and NKA as well.
IV. CENTRAL MOVEMENT DISORDERS
The mammalian TK system has been implicated in several centrally-based neurological movement disorders that share the commonality of nigrostriatal degeneration. SP and NKB are both are expressed in D1R-containing striatal neurons [36–39]. SP is co-localized with dynorphin and D1Rs in striatonigral neurons [38], whereas NKB is co-localized with D1Rs in a unique population of striatal projection neurons [39, 40]. TAC1Rs are highly expressed in many brain regions involved in motor control, including dopaminergic regions of the striatum [41, 42]. TACR3 receptors are moderately expressed in the brain but are concentrated in GABA interneurons in striatum and in dopamine neurons of substantia nigra [43–46]. Therefore, it is not surprising that pharmacological TK-system targeting of central dopaminergic disorders is gaining interest [47–49].
Parkinson’s disease (PD) is a neurodegenerative motor disorder characterized by rigidity, bradykinesia, and resting tremor. The primary cause of PD is a degeneration of dopamine neurons within the substantia nigra and basal ganglia. Multiple studies have uncovered a correlation between the onset of PD and the degeneration of neurons containing SP throughout the substantia nigra and basal ganglia, as well as in surrounding areas. The human pedunculopontine tegmental nucleus (PPTN) is associated with movement [50], and neurons within the PPTN have been shown to contain SP [51]. Analysis of SP-containing neurons within the PPTN (as well as the laterodorsal tegmental nucleus and the oral pontine reticular nucleus) of deceased subjects with no apparent neurological disease showed that subjects diagnosed post-mortem with PD had severely damaged SP-positive neurons. It was also shown that the older post-mortem PD patients possessed the least total SP-positive neurons in the mesopontine tegmentum [52], suggesting SP neuron-specific progressive degeneration. Similar findings have been reported regarding the status of SP-positive neurons in the dorsal motor vagal nuclei [51], the globus pallidus [53], and the substantia nigra [53, 54] of human post-mortem PD brains. In an attempt to elucidate the role of SP in PD, Cui and colleagues examined the involvement of the pallidal SP system in PD by observing the electrophysiological effects of SP on pallidal neurons of 6-OHDA-lesioned rats [55]. Neurons in the globus pallidus exposed to SP demonstrated a smaller increase in firing rate in 6-OHDA-treated rats than in control rats, indicating a decreased expression of functional TACR1s in the globus pallidus in the PD model. They also found that a selective TACR1 antagonist blocked SP-induced excitation, suggesting the involvement of TACR1 receptors in modulating electrical activity in the globus pallidus of PD patients [55]. The 6-OHDA-lesion rat model has also been used to analyze the role of NKB-TACR3 system in PD [56, 57]. After 6-OHDA lesioning, NKB mRNA expression was increased and could be even further enhanced after the lesion by repeated L-DOPA treatment. In contrast to NKB levels, the expression of SP mRNA decreased after the lesion [57]. From this study, it is apparent that in contrast to SP, dopamine regulates NKB mRNA in a bidirectional manner [57]. The potential to develop TK-based therapeutics to treat PD has not been overlooked. In fact, numerous studies have reported DA neuron growth in response to intracerebroventricular (i.c.v.) administration of SP [58], as well as TACR1 and TACR3 agonists [59].
Machado-Joseph disease (MJD), also known as Nigrospinodental Degeneration (ND) or spinocerebellar ataxia type 3 (SCA3), is a neurodegenerative disease with symptoms reminiscent of PD [60]. MJD is the most common type of autosomal dominantly-inherited cerebellar ataxias in Europe, Japan, and the United States [61]. MJD is believed to be caused by a CAG expansion in the SCA3 gene located on chromosome 14q32.1 and is characterized by poor motor control. Based on previous anatomic, biochemical, and pharmacological studies associating SP with dopaminergic neurons in the substantia nigra and the striatum [62–64], Matsuishi and colleagues conducted a study comparing the CSF SP levels of 7 subjects with MJD to 14 controls. They found that the level of SP within the CSF of patients with MJD was significantly lower than the levels observed in the control patients. Despite these promising findings, however, no correlations were found between the CSF level of SP and the presence or absence of clinical symptoms such as disease duration, severity, type, or treatment, or clinical symptoms of peripheral nerve involvement or signs of autonomic dysfunction [65]. The authors posited that these findings could reflect a decreased concentration of SP in the neurons, which may be indicative of the involvement of the peripheral nerves or the dorsal root ganglia, which is characteristic of MJD.
V. TRAUMATIC BRAIN INJURY
Recent studies have suggested that SP, as well as a number of other neuropeptides, may play a substantial role in the functional and morphological degeneration following acute injury in both the peripheral nervous system (PNS) and CNS [66]. Acute traumatic brain injury (TBI) results in the generation of neurologic deficits through two primary mechanisms [67]. The primary injury occurs at the time of the trauma and includes mechanical processes, such as shearing, laceration, and stretching of nerve fibers. Secondary injury occurs at later time points and is composed of biochemical and physiological factors that are initiated by the primary event and manifest over time [67]. Morbidity after brain injury is strongly associated with the development of this secondary injury cascade. Secondary injury factors include blood–brain barrier rupture, edema, release of excitatory amino acids, disrupted ion homeostasis, oxidative stress, and metabolic failure [67–69]. It is well known that edema is highly related to outcome after injury; however, the mechanisms associated with the formation of edema are still unclear. With respect to peripheral tissue edema, an association between neuropeptides and the development of increased vascular permeability and edema is evident and has been generically termed ‘neurogenic inflammation’ [70]. Neurogenic inflammation is a neurally elicited reaction that involves vasodilation, increased microvascular permeability, protein extravasation, and tissue swelling. Studies of peripheral nerves have shown that neurogenic inflammation is the result of stimulation of C-fibers, which express several neuropeptides, including SP and NKA [71, 72]. Trauma-induced, uncontrolled edema formation in the CNS elevates intracranial pressure, which can cause disruption of cerebral blood flow. This vascular compression can lead to reduced perfusion, localized hypoxia and ischemia, and eventual neuronal death. Therefore, it is of particular interest to pharmacologically attenuate or inhibit such inflammation after TBI. Malcangio and colleagues have demonstrated that SP is released in the nervous system following peripheral nerve injury (by measuring SP expression in axotomized large afferents) and is downregulated in small fibers following stimulation of Aβ-fibers and Aβ/Aδ/C-fibers [73]. SP release in the thoracic spinal cord has also been directly measured following focal traumatic injury [74]. Perivascular SP immunoreactivity has been shown to increase following TBI, and this was irrespective of the severity of the injury and occurred in both focal and diffuse injury models [75]. Therefore, one of the potential therapeutic mechanisms that could be exploited for the treatment of TBI would be receptor antagonism directly after the initial trauma [76–78]. These therapeutic endeavors will be discussed in greater detail later in this review.
VI. ALZHEIMER’S DISEASE
Alzheimer’s disease (AD) is a common form of pre-senile dementia that is dominantly characterized by progressive memory loss, as well as cognitive and linguistic impairments. AD is one of most prevalent diseases in Westernized countries, and an efficacious treatment strategy represents one of the most important challenges in modern medicine [79, 80]. The physiological mechanisms leading to the generation of AD pathophysiology are complex and multifactorial [81–83]. However, there is a general consensus that post-translational alterations in the cytoskeletal protein tau as well as changes in amyloid precursor protein processing are pivotal. The primary sites of these molecular pathologies are the hippocampus and cortex, although other brain regions can be subsequently affected. Although a correlation between TKs, in particular SP, and AD is well documented [84, 85], most studies indicate that SP is not a causal factor in the development of AD. Several studies have documented a decrease in SP immunoreactive tissue in various regions of AD brains, e.g., significant reductions in SP-positive cells are found in the midfrontal, inferior parietal, and occipital lobes, as well as the superior and inferior temporal gyri [86]. Similar studies have shown decreased SP-positive cells in the frontal and occipital cortices, as well as the isocortex, hippocampus, and to a lesser extent, the allocortex [84, 87–89]. In addition, substance P-immunoreactive profiles coexist within single senile amyloid-containing plaques of patients with AD [90]. Despite parallels between AD pathology and the presence of SP, there is a relative scarcity of evidence that SP plays a prominent role in the development of AD [91, 92]. In a study of five distinct CSF biomarkers, no significant difference between AD and healthy patients with respect to SP immunoreactivity has been observed [93]. However, within AD subjects, patients with late onset (>65 years old) AD had significantly higher levels of SP than both patients with earlier onset AD and control patients [93].
With respect to a potential mechanistic connection between SP and cognitive function, it has been demonstrated that expression of SP, NKB, and the TACR1 is especially high in brain regions critical for the regulation of emotion and cognition [94, 95]. The SP ligand system functionally interacts with the cholinergic ascending system of the nucleus basalis Meynert, which is one of the main neuronal pathway deficiencies observed in AD. Recreation of the AD pathology with ibotenic acid infusion into the rat nucleus basalis magnocellularis, can induce significant reductions in SP levels in the frontal cortex and striatum [96]. Conversely, injection of SP into the nucleus basalis magnocellularis has been shown to increase acetylcholine levels in the hippocampus and cortex of rats in a TACR1-dependent manner [97, 98].
VII. HUNTINGTON’S DISEASE
Huntington’s disease (HD) is a genetically linked neurodegenerative disorder first described by George Huntington in 1872. It is a hyperkinetic disorder specifically characterized by autosomally-dominant heritability; presentation of chorea or jerky, dance-like movements; behavioral and physical disturbances, including personality changes and skewed balance; cognitive impairments, such as depression or dementia; and death 10–15 years after initial onset [99, 100]. HD is equally frequent in men and women, and its incidence in the whole population is approximately 5–10 cases per 100,000 worldwide, making it the most common inherited neurodegenerative disorder [101]. The primary cause of the disorder was identified as an expanded trinucleotide repeat (CAG) in the gene encoding for the huntingtin protein (htt). It is interesting to note, however, that in addition to the archetypical neurological phenotypic attributes, many HD patients develop a specific form of genetic diabetes and experience extreme weight loss, which is mirrored in animal models of the genetic disorder [102].
As with several other neurodegenerative disorders, SP is generally found to be decreased in subjects diagnosed with HD. HD is generally characterized by neuronal loss in the striatum [103], including severe losses in the dorsal caudate nucleus and putamen [104, 105]. Multiple studies have addressed the loss of SP-positive neurons throughout the striatum and other regions of the basal ganglia, including the internal pallidal segment of the globus pallidus and the substantia nigra [106]. In a study of the differential sparing of compartments in the striatum of HD patients, as related to the distribution of SP, Ferrante and colleagues found a dorsoventral gradient of SP immunoreactivity loss in the striatum that was directly correlated with the severity of the disease. The most severe cases of HD showed very little to no staining for SP in the caudate nucleus and dorsal putamen, whereas the absence of SP in cases of moderately severe HD was less extensive [104]. This loss of SP indicated a reduction of intrinsic spiny neurons which project to the globus pallidus and substantia nigra [103], which are known to be preferentially affected in HD [107]. Additionally, a correlation between classical HD pathology and TK levels has also been demonstrated in an HD-related condition, i.e., ‘HD phenocopy’. Phenocopies of HD are individuals with a family history, clinical symptoms, and occasionally pathological evidence of HD who do not possess the expanded CAG repeat within the huntingtin (htt) gene. HD phenocopy patients often present with selective loss of preprotachykinin neurons and dysfunction of surviving preprotachykinin neurons within the striatum. In addition to this disruption, HD phenocopies also demonstrate a profound loss of immunohistochemical staining for SP in terminals of striatal neurons projecting to the substantia nigra [106].
TACHYKININ NEUROTHERAPEUTIC MECHANISMS
As we have seen, there is considerable evidence linking alterations of the TK ligand and receptor system in multiple central neurological disorders. It is clear that with respect to these diseases, manipulation of the TK receptor-ligand system may prove beneficial either by stemming the progression of the disorders or through a direct therapeutic mechanism. In the next section of this review, we will delineate the molecular and neurophysiological mechanisms by which modulation of the TK receptor-ligand system may yield significant therapeutics actions. These mechanisms include actions that can repair, protect, or even replace neural tissue damaged either by congenital disease or trauma.
I. NEUROTROPHIC MECHANISMS
The ability of SP to promote neuronal growth is a well-documented phenomenon. In recent times, the concept of adult neurogenesis has been promoted as a potential mechanism by which damaged neuronal tissue can be replaced by activation and development of neural progenitor cells. In a study designed to measure the effects of SP on the rat cortical subventricular zone (SVZ) and the hippocampal dentate gyrus (DG), which are areas known to possess a neuronal stem cell population, it was found that with the introduction of SP, there was a significant increase in the number of neural progenitor cells (NPCs) in the rat SVZ and DG [108]. Central nervous system neurogenesis has also been studied in TACR1(−/−) mice. These mice demonstrate significantly elevated levels of neurogenesis in the DG compared to control mice [109]. This increase in neurogenesis, however, seems to be strain-dependent, as genetic ablation of TACR1 in a B6 strain failed to potentiate adult neurogenesis, while in a 129B6 strain background, the increase in neurogenesis [109] was replicated [110]. These two strains differed in the level of plasma corticosterone, suggesting a potential link between the TK-control of neurogenesis and stress-response mechanisms. With both ligand stimulation and receptor expression levels controlling NPC levels in a complicated manner, it seems likely that the future manipulation of the TK system may be employed to control the replacement or the rejuvenation of central nervous tissue.
II. NEUROINFLAMMATION
Inflammatory responses within the CNS are often the result of traumatic injury and infectious agents [111]. Inflammation, while often representative of a protective immune response, may also result in progressive damage to the CNS. There is significant evidence to indicate that SP is a major component of the inflammatory responses at peripheral sites, as well as CNS cell types. Therefore, the ability to specifically control TK-related activities in central inflammatory responses may represent an important pharmacotherapeutic mechanism for centrally acting TK drugs. Numerous studies have shown that astrocyte-derived SP may activate several transcriptional regulators that play an important role in the regulation of pro-inflammatory molecule expression. For example, NF-κB [112, 113] and p38 MAP kinase [113] have both been shown to be activated by SP in central nervous tissue. Both SP and its cognate TACR1 receptor are also known to be expressed at high levels in murine microglia [114, 115] and are thought to play a role via NF-κB activation in the production of the pro-inflammatory cytokines TNF-alpha, IL-1, and IL-6 [116–118]. In addition to the neuroinflammatory-promoting properties of SP, NKA has also been associated with the inflammatory cytokines IL-1 [119] and IL-6 [120]. It is important to note, however, that some studies suggest that SP alone is not sufficient to elicit neuroinflammatory immune responses and that it must work with other microglial responses to inflammatory stimuli [116, 121].
III. NEUROPROTECTION
Neuronal tissue can be severely damaged either through physical trauma or excitotoxic insults. Both of these processes can result in calcium overload, protein degradation, or DNA damage. There are endogenous cellular responses that can be activated by neuronal tissue in response to damage that attempt to protect cellular, protein, and nucleic acid integrity. There is a large variety of endogenous neuroprotective signaling mechanisms that can be mimicked by pharmacotherpeutics to ameliorate CNS insults. As with many forms of activity of neuromodulators in the CNS, there are instances in which TKs have been implicated as players in a number of physiological and pathological conditions (e.g., the pro-convulsant role of the PPTA gene [35] and the involvement of SP in TBI [73–75]), but there are also numerous illustrations of the neuroprotective qualities of TKs both in vitro and in vivo. For example, multiple TK agonists are able to protect cerebellar granule cells from neuronal cell death induced by serum and potassium deprivation. These neuroprotective effects of SP have been found to be mediated by activation of the kinases Akt-1 and extracellular signal-regulated kinase (ERK) [122]. Similar protective actions of TKs acting at the TACR1 have been seen in dissociated spiral ganglia when they were exposed to trophic factor deprivation shock [123]. In contrast to the activation of protective kinases, such as Akt-1, the neuroprotective action of TK was mediated via a TACR1-dependent inhibition of pro-apoptotic caspase enzymes. While many studies have concentrated on the neuroprotective abilities of SP, other mammalian TKs also display such beneficial activity. Hence, when SP was unable to protect striatal cholinergic neurons, NKA and NKB were more potent at protecting the neuronal cells from an excitotoxic (kainic acid) insult [124]. As cholinergic pathways have been demonstrated to play a crucial role in cognitive decline and AD, it is important to note that SP was recently demonstrated to protect cerebellar granule neurons from amyloid beta (Aβ)-mediated cytotoxicity [125]. This action of SP was thought to occur via an SP-mediated downregulation and functional inhibition of specific voltage-gated potassium channels. When tested in a model in which dopaminergic (DA) neurons die spontaneously and progressively as a function of time, SP and NKB were all shown to protect and maintain the numbers of tyrosine hydroxylase-labeled neurons in mesencephalic cultures [59].
NEUROTHERAPEUTIC APPLICATIONS OF TK-SYSTEM LIGANDS
As we have outlined in this review, it is clear that the TK receptor-ligand system can have profound effects on multiple neurological disorders, including amyotrophic lateral sclerosis, Alzheimer’s disease, Parkinson’s disease, traumatic brain injury and central motor disorders. The neuroprotective nature of tachykinins makes TK agonists prime candidates for therapeutic agents in neurodegenerative conditions. A vital aspect in the treatment of Parkinson’s disease is the prevention of dopaminergic cell death. Multiple TK agonists have been demonstrated to prevent the progressive loss of dopaminergic cells in models of nigrostriatal degeneration [58, 59]. With regard to Alzheimer’s disease, several studies have indicated an extension of the neuroprotective attributes of SP into the realm of neuronal death induced by amyloid-β (Aβ). In the early 1990s, several research teams demonstrated that TK peptide agonists were able to nearly completely reverse the trophic and toxic effects of Aβ [126, 127].
TK receptor antagonists, specifically TACR1 antagonists, have been especially successful in clinical settings when treating nausea and vomiting [128]. TACR1 antagonists have also demonstrated promise for affective disorders linked with stress and anxiety. Multiple TACR1 antagonists have been shown to attenuate the anxiety-inducing effects of centrally-infused SP agonists [129, 130] and later proved to be as effective as paroxetine, a selective serotonin reuptake inhibitor, in treating major depressive disorder in humans [129, 131]. There have also been a number of studies investigating the treatment of chemotherapy-induced emesis with MK-869, as well as other TACR1 antagonists. However, MK-869 ameliorates nausea only when taken in tandem with a serotonin 5-HT3 receptor antagonist and dexamethasone, a known antiemetic [132]. Although research into possible treatment options of TBI using TK receptor antagonists is not as developed as it is in other fields, there are several indicators that point to TACR1 antagonists as a potential therapeutic avenue. TACR1 antagonists have been found to reduce post-ischemic myocardial injury in rats deprived of dietary magnesium, primarily through a potential anti-inflammatory action [133]. Traumatic injury has been shown to produce sustained decline in intracellular magnesium, suggesting that SP may contribute to the detrimental effects of magnesium deficiency [134]. Currently there are two TACR1 antagonists (Aprepipant [129] and Fosaprepipant) marketed for their ability to ameliorate nausea symptomology. However, there are also TACR1, TACR2, and TACR3 antagonists in various phases of clinical trials. Antagonists for the TACR1 are being assessed for addiction/post-traumatic stress disorder (Aprepipant, Vofopitant), major depressive disorder (Orvepitant), seasonal affective disorder (LY-686017, Vestipitant), and schizophrenia (AZD-2624). Antagonists for the TACR2 or TACR3 are also currently under investigation in clinical trials for major depressive disorder (TACR2: Saredutant), acute panic attacks (TACR3: Osanetant), and schizophrenia (TACR3: Talnetant, SSR-241586).
CONCLUSIONS
The extensive involvement of tachykinins (TKs) and their receptors in a wide range of biological functions has made them prime targets for physiological and pharmacological investigation. These peptides seem to display great diversity in their involvement with a number of CNS disorders, ranging from minor roles in ALS and MJD to significant contributions in affective and neurodegenerative disorders. The sheer breadth of the associations between TKs and multiple CNS disorders is likely to lead, via advanced pharmacological targeting [80, 135], to even more TK-based neurotherapeutics. Further investigation of the complex interactions of the TK receptor-ligand system with other neurotransmitter systems will undoubtedly increase the potential impact of novel pharmacotherapeutics targeted towards these TK receptors.
Acknowledgments
This research was supported by the Intramural Research Program of the NIH, National Institute on Aging.
ABBREVIATIONS
- AD
Alzheimer’s disease
- ALS
amyotrophic lateral sclerosis
- CNS
central nervous system
- CSF
cerebrospinal fluid
- GPCR
G protein-coupled receptor
- HD
Huntington’s disease
- MJD
Machado-Joseph disease
- NKA
neurokinin A
- NKB
neurokinin B
- PD
Parkinson’s disease
- SP
substance P
- TBI
traumatic brain injury
- TK
tachykinin
Footnotes
The authors have no conflicts of scientific interest with respect to the manuscript.
References
- 1.Tatemoto K, Lundberg JM, Jörnvall H, Mutt V. Neuropeptide K: isolation, structure and biological activities of a novel brain tachykinin. Biochem Biophys Res Commun. 1985;128(2):947–953. doi: 10.1016/0006-291x(85)90138-x. [DOI] [PubMed] [Google Scholar]
- 2.Kage R, McGregor GP, Thim L, Conlon JM. Neuropeptide-gamma: a peptide isolated from rabbit intestine that is derived from gamma-preprotachykinin. J Neurochem. 1988;50(5):1412–1417. doi: 10.1111/j.1471-4159.1988.tb03024.x. [DOI] [PubMed] [Google Scholar]
- 3.Kurtz MM, Wang R, Clements MK, Cascieri MA, Austin CP, Cunningham BR, Chicchi GG, Liu Q. Identification, localization and receptor characterization of novel mammalian substance P-like peptides. Gene. 2002;296(1–2):205–212. doi: 10.1016/s0378-1119(02)00861-2. [DOI] [PubMed] [Google Scholar]
- 4.Von Euler US, Gaddum JH. An unidentified depressor substance in certain tissue extracts. J Physiol. 1931;72(1):74–87. doi: 10.1113/jphysiol.1931.sp002763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Erspamer V, Espamer GF. Pharmacological actions of eledoisin on extravascular smooth muscle. Br J Pharmacol. 1962;19(2):337–354. doi: 10.1111/j.1476-5381.1962.tb01198.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Chang MM, Leeman SE, Niall HD. Amino-acid sequence of substance P. Nat New Biol. 1971;232(29):86–87. doi: 10.1038/newbio232086a0. [DOI] [PubMed] [Google Scholar]
- 7.Kangawa K, Minamino N, Fukuda A, Matsuo H. Neuromedin K: a novel mammalian tachykinin identified in porcine spinal cord. Biochem Biophys Res Commun. 1983;114(2):533–540. doi: 10.1016/0006-291x(83)90813-6. [DOI] [PubMed] [Google Scholar]
- 8.Kimura S, Okada M, Sugita Y, Kanazawa I, Munekate E. Novel neuropeptides, neurokinin α and β, isolated from porcine spinal cord. Proc Jpn Acad B. 1983;59(1):101–104. [Google Scholar]
- 9.Regoli S, Boudon A, Fauchére JL. Receptors and antagonists for substance P and related peptides. Pharmacol Rev. 1994;46(4):551–599. [PubMed] [Google Scholar]
- 10.Zhang Y, Lu L, Furlonger C, Wu GE, Paige CJ. Hemokinin is a hematopoietic-specific tachykinin that regulates B lymphopoiesis. Nat Immunol. 2000;1(5):392–397. doi: 10.1038/80826. [DOI] [PubMed] [Google Scholar]
- 11.Almeida TA, Rojo J, Nieto PM, Pinto FM, Hernandez M, Martín JD, Candenas ML. Tachykinins and tachykinin receptors: structure and activity relationships. Curr Med Chem. 2004;11(15):2045–2081. doi: 10.2174/0929867043364748. [DOI] [PubMed] [Google Scholar]
- 12.Ohkubo H, Nakanishi S. Molecular characterization of the three tachykinin receptors. Ann N Y Acad Sci. 1991;632:53–62. doi: 10.1111/j.1749-6632.1991.tb33094.x. [DOI] [PubMed] [Google Scholar]
- 13.Maggi CA. The mammalian tachykinin receptors. Gen Pharmacol. 1995;26(5):911–944. doi: 10.1016/0306-3623(94)00292-u. [DOI] [PubMed] [Google Scholar]
- 14.Maudsley S, Carpenter E, Gent JP. Receptor characterization of the NK1 receptor on NG108–15 cells. Br J Pharmacol. 1993;110:175. [Google Scholar]
- 15.Gerard NP, Bao L, Xiao-Ping H, Gerard C. Molecular aspects of the tachykinin receptors. Regul Pept. 1993;43(1–2):21–35. doi: 10.1016/0167-0115(93)90404-v. [DOI] [PubMed] [Google Scholar]
- 16.Maudsley S, Gent JP. Tachykinin-induced activation of a non-selective cation conductance in ND7/23 cells. Br J Pharmacol. 1995;116:444. [Google Scholar]
- 17.Phenna S, Carpenter E, Peers C, Maudsley S, Gent JP. Inhibition of Ca2+-sensitive K+ currents in NG108–15 cells by substance P and related tachykinins. Br J Pharmacol. 1996;119:315–320. doi: 10.1111/j.1476-5381.1996.tb15988.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Maudsley S, Gent JP, Findlay JB, Donnelly D. The relationship between the agonist-induced activation and desensitization of the human tachykinin NK2 receptor expressed in Xenopus oocytes. Br J Pharmacol. 1998;124(4):675–684. doi: 10.1038/sj.bjp.0701889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Mantyh PW. Neurobiology of substance P and the NK1 receptor. J Clin Psychiatry. 2002;63(11):6–10. [PubMed] [Google Scholar]
- 20.Herpfer I, Lieb K. Substance P and Substance P receptor antagonists in the pathogenesis and treatment of affective disorders. World J Biol Psychiatry. 2003;4(2):56–63. doi: 10.3109/15622970309167952. [DOI] [PubMed] [Google Scholar]
- 21.Schedlowski M, Fluge T, Richter S, Tewes U, Schmidt RE, Wagner TO. Beta-endorphin, but not substance-P, is increased by acute stress in humans. Psychoneuroendocrinology. 1995;20(1):103–110. doi: 10.1016/0306-4530(94)00048-4. [DOI] [PubMed] [Google Scholar]
- 22.Weiss DW, Hirt R, Tarcic N, Berzon Y, Ben-Zur H, Breznitz S, Glaser B, Grover NB, Baras M, O’Dorisio TM. Studies in psychoneuroimmunology: psychological, immunological, and neuroendocrinological parameters in Israeli civilians during and after a period of Scud missile attacks. Behav Med. 1996;22(1):5–14. doi: 10.1080/08964289.1996.9933760. [DOI] [PubMed] [Google Scholar]
- 23.Bondy B, Baghai TC, Minov C, Schüle C, Schwarz MJ, Zwanzger P, Rupprecht R, Möller HJ. Substance P serum levels are increased in major depression: preliminary results. Biol Psychiatry. 2003;53(6):538–42. doi: 10.1016/s0006-3223(02)01544-5. [DOI] [PubMed] [Google Scholar]
- 24.Rimon R, Le Greves P, Nyberg F, Heikkila L, Salmela L, Terenius L. Elevation of substance P-like peptides in the CSF of psychiatric patients. Biol Phychiatry. 1984;19(4):509–516. [PubMed] [Google Scholar]
- 25.Vaeroy H, Helle R, Forre O, Kass E, Terenius L. Elevated CSF levels of substance P and high incidence of Raynaud phenomenon in patients with fibromyalgia: new features for diagnosis. Pain. 1988;32(1):21–26. doi: 10.1016/0304-3959(88)90019-X. [DOI] [PubMed] [Google Scholar]
- 26.Russell IJ, Orr MD, Littman B, Vipraio CA, Alboukrek D, Michalek JE, Lopez Y, MacKillip F. Elevated cerebrospinal fluid levels of substance P in patients with the fibromyalgia syndrome. Arthritis Rheum. 1994;37(11):1593–1601. doi: 10.1002/art.1780371106. [DOI] [PubMed] [Google Scholar]
- 27.Russell IJ. The promise of substance P inhibitors in fibromyalgia. Rheum Dis Clin N Am. 2002;28(2):329–342. doi: 10.1016/s0889-857x(01)00006-0. [DOI] [PubMed] [Google Scholar]
- 28.Pae CU, Marks DM, Patkar AA, Masand PS, Luyten P, Serretti A. Pharmacological treatment of chronic fatigue syndrome: focusing on the role of antidepressants. Expert Opin Pharmacother. 2009;10(10):561–570. doi: 10.1517/14656560902988510. [DOI] [PubMed] [Google Scholar]
- 29.Lieb K, Ahlvers K, Dancker K, Strohbusch S, Reincke M, Feige B, Berger M, Riemann D, Voderholzer U. Effects of the neuropeptide substance P on sleep, mood, and neuroendocrine measures in healthy young men. Neuropsychopharmacol. 2002;27(6):1041–1049. doi: 10.1016/S0893-133X(02)00369-X. [DOI] [PubMed] [Google Scholar]
- 30.Gillberg PG, Aquilonius SM, Eckernäs SA, Lundqvist G, Winblad B. Choline acetyltransferase and substance P-like immuno-reactivity in the human spinal cord: changes in amyotrophic lateral sclerosis. Brain Res. 1982;250(2):394–397. doi: 10.1016/0006-8993(82)90439-5. [DOI] [PubMed] [Google Scholar]
- 31.Matsuishi T, Nagamitsu S, Shoji H, Itoh M, Takashima S, Iwaki T, Shida N, Yamashita Y, Sakai T, Kato H. Increased cerebrospinal fluid levels of substance P in patients with amyotrophic lateral sclerosis. Short communication. J Neural Transm. 1999;106(9–10):943–948. doi: 10.1007/s007020050214. [DOI] [PubMed] [Google Scholar]
- 32.Ito H, Kusaka H, Matsumoto S, Imai T. Topographic involvement of the striatal efferents in basal ganglia of patients with adult-onset motor neuron disease with basophilic inclusions. Acta Neuropathol. 1995;89(6):513–518. doi: 10.1007/BF00571505. [DOI] [PubMed] [Google Scholar]
- 33.Zachrisson O, Lindefors N, Brene S. A tachykinin NK1 receptor antagonist, CP-122,721–1, attenuates kainic acid-induced seizure activity. Mol Brain Res. 1998;60(2):291–295. doi: 10.1016/s0169-328x(98)00191-0. [DOI] [PubMed] [Google Scholar]
- 34.Liu H, Mazarati AM, Katsumori H, Sankar R, Wasterlain CG. Substance P is expressed in hippocampal principal neurons during status epilepticus and plays a critical role in the maintenance of status epilepticus. Proc Natl Acad Sci. 1999;96(9):5286–5291. doi: 10.1073/pnas.96.9.5286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Liu H, Cao Y, Basbaum AI, Mazarati AM, Sankar R, Wasterlain CG. Resistance to excitotoxin-induced seizures and neuronal death in mice lacking the preprotachykinin A gene. Pro Natl Acad Sci. 1999;96(21):12096–12101. doi: 10.1073/pnas.96.21.12096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Bonner TI, Affolter HU, Young AC, Young WS., 3rd A cDNA encoding the precursor of the rat neuropeptide, neurokinin B. Brain Res. 1987;388:243–249. doi: 10.1016/0169-328x(87)90031-3. [DOI] [PubMed] [Google Scholar]
- 37.Krause JE, Chirgwin JM, Carter MS, Xu ZS, Hershey AD. Three rat preprotachykinin mRNAs encode the neuropeptides substance P and neurokinin A. Proc Natl Acad Sci US A. 1987;84:881–885. doi: 10.1073/pnas.84.3.881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Gerfen CR, Engber TM, Mahan LC, Susel Z, Chase TN, Monsma FJ, Jr, Sibley DR. D1 and D2 dopamine receptor-regulated gene expression of striatonigral and striatopallidal neurons. Science. 1990;250(4986):1429–1432. doi: 10.1126/science.2147780. [DOI] [PubMed] [Google Scholar]
- 39.Sonomura T, Nakamura K, Furuta T, Hioki H, Nishi A, Yamanaka A, Uemura M, Kaneko T. Expression of D1 but not D2 dopamine receptors in striatal neurons producing neurokinin B in rats. Eur J Neurosci. 2007;26:3093–3103. doi: 10.1111/j.1460-9568.2007.05923.x. [DOI] [PubMed] [Google Scholar]
- 40.Furuta T, Mori T, Lee T, Kaneko T. Third group of neostriatofugal neurons: neurokinin B-producing neurons that send axons predominantly to the substantia innominata. J Comp Neurol. 2000;426:279–296. doi: 10.1002/1096-9861(20001016)426:2<279::aid-cne9>3.0.co;2-f. [DOI] [PubMed] [Google Scholar]
- 41.Sivam SP, Krause JE. Tachykinin systems in the spinal cord and basal ganglia: influence of neonatal capsaicin treatment or dopaminergic intervention on levels of peptides. substance P-encoding mRNAs, and substance P receptor mRNA. J Neurochem. 1992;59:2278–84. doi: 10.1111/j.1471-4159.1992.tb10121.x. [DOI] [PubMed] [Google Scholar]
- 42.Whitty CJ, Paul MA, Bannon MJ. Neurokinin receptor mRNA localization in human midbrain dopamine neurons. J Comp Neurol. 1997;382(3):394–400. [PubMed] [Google Scholar]
- 43.Shughrue PJ, Lane MV, Merchenthaler I. In situ hybridization analysis of the distribution of neurokinin-3 mRNA in the rat central nervous system. J Comp Neurol. 1996;372:395–314. doi: 10.1002/(SICI)1096-9861(19960826)372:3<395::AID-CNE5>3.0.CO;2-Y. [DOI] [PubMed] [Google Scholar]
- 44.Chen LW, Guan ZL, Ding YQ. Mesencephalic dopaminergic neurons expressing neuromedin K receptor (NK3): a double immunocytochemical study in the rat. Brain Res. 1998;780:150–154. [PubMed] [Google Scholar]
- 45.Preston Z, Richardson PJ, Pinnock RD, Lee K. NK-3 receptors are expressed on mouse striatal gamma-aminobutyric acid-ergic interneurones and evoke [(3)H] gamma-aminobutyric acid release. Neurosci Lett. 2000;284:89–92. doi: 10.1016/s0304-3940(00)00968-x. [DOI] [PubMed] [Google Scholar]
- 46.Furuta T, Koyano K, Tomioka R, Yanagawa Y, Kaneko T. GABAergic basal forebrain neurons that express receptor for neurokinin B and send axons to the cerebral cortex. J Comp Neurol. 2004;473:43–58. doi: 10.1002/cne.20087. [DOI] [PubMed] [Google Scholar]
- 47.Dawson LA, Cato KJ, Scott C, Watson JM, Wood MD, Foxton R, de la Flor R, Jones GA, Kew JN, Cluderay JE, Southam E, Murkitt GS, Gartlon J, Pemberton DJ, Jones DN, Davies CH, Hagan J. In vitro and in vivo characterization of the non-peptide NK3 receptor antagonist SB-223412 (talnetant): potential therapeutic utility in the treatment of schizophrenia. Neuropsychopharmacology. 2008;33(7):1642–1652. doi: 10.1038/sj.npp.1301549. [DOI] [PubMed] [Google Scholar]
- 48.Spooren W, Riemer C, Meltzer H. NK3 receptor antagonists: the next generation of antipsychotics? Nat Rev Drug Discov. 2005;4:967–75. doi: 10.1038/nrd1905. [DOI] [PubMed] [Google Scholar]
- 49.Chen LW, Yung KK, Chan YS. Neurokinin peptides and neurokinin receptors as potential therapeutic intervention targets of basal ganglia in the prevention and treatment of Parkinson’s disease. Curr Drug Targets. 2004;5(2):197–206. doi: 10.2174/1389450043490596. [DOI] [PubMed] [Google Scholar]
- 50.Garcia-Rill E, Huser CR, Skiner RD, Smith W, Woodward DJ. Locomotion-inducing sites in the vicinity of the pedunculopontine nucleus. Brain Res Bul. 1987;18(6):731–738. doi: 10.1016/0361-9230(87)90208-5. [DOI] [PubMed] [Google Scholar]
- 51.Halliday GM, Gai WP, Blessing WW, Geffen LB. Substance P-containing neurons in the pontomesencephalic tegmentum of the human brain. Neurosci. 1990;39(1):81–96. doi: 10.1016/0306-4522(90)90223-q. [DOI] [PubMed] [Google Scholar]
- 52.Gai WP, Halliday GM, Blumbergs PC, Geffen LB, Blessing WW. Substance P-containing neurons in the mesopontine tegmentum are severely affected in Parkinson’s disease. Brain. 1991;114(5):2253–2267. doi: 10.1093/brain/114.5.2253. [DOI] [PubMed] [Google Scholar]
- 53.Mauborgne A, Javoy-Agid F, Legrand JC, Agid Y, Cesselin F. Decrease of substance P-like immunoreactivity in the substantia nigra and pallidum of parkinsonian brains. Brain Res. 1983;268(1):167–170. doi: 10.1016/0006-8993(83)90403-1. [DOI] [PubMed] [Google Scholar]
- 54.Tenovuo O, Rinne UK, Viljanen MK. Substance P immunoreactivity in the post-mortem parkinsonian brain. Brain Res. 1984;303(1):113–116. doi: 10.1016/0006-8993(84)90217-8. [DOI] [PubMed] [Google Scholar]
- 55.Cui Q, Yung W, Chen L. Effects of substance P on neuronal firing of pallidal neurons in parkinsonian rats. Neurosci Res. 2008;60(2):162–169. doi: 10.1016/j.neures.2007.10.007. [DOI] [PubMed] [Google Scholar]
- 56.Burgunder JM, Young WS., 3rd Distribution, projection and dopaminergic regulation of the neurokinin B mRNA-containing neurons of the rat caudate-putamen. Neurosci. 1989;32(2):323–335. doi: 10.1016/0306-4522(89)90081-x. [DOI] [PubMed] [Google Scholar]
- 57.Zhang X, Andren PE, Chergui K, Svenningsson P. Neurokinin B/NK3 receptors exert feedback inhibition on L-DOPA actions in the 6-OHDA lesion rat model of Parkinson’s disease. Neuropharma. 2008;54(7):1143–1153. doi: 10.1016/j.neuropharm.2008.03.005. [DOI] [PubMed] [Google Scholar]
- 58.Krasnova IN, Bychkov ER, Lioudyno VI, Zubareva OE, Dambinova SA. Intracerebroventricular administration of substance P increases dopamine content in the brain of 6-hydroxydopamine-lesioned rats. Neurosci. 2000;95(1):113–117. doi: 10.1016/s0306-4522(99)00400-5. [DOI] [PubMed] [Google Scholar]
- 59.Salthun-Lassalle B, Traver S, Hirsch EC, Michel PP. Substance P, neurokinins A and B, and synthetic tachykinin peptides protect mesencephalic dopaminergic neurons in culture via an activity-dependent mechanism. Mol Pharma. 2005;68(5):1214–1224. doi: 10.1124/mol.105.015453. [DOI] [PubMed] [Google Scholar]
- 60.Schols L, Vieira-Saecker AM, Schols S, Przuntek H, Epplen JT, Riess O. Trinucleotide expansion within the MJD1 gene presents clinically as spinocerebellar ataxia and occurs most frequently in German SCA patients. Hum Mol Genet. 1995;4:1001–1005. doi: 10.1093/hmg/4.6.1001. [DOI] [PubMed] [Google Scholar]
- 61.Schols L, Bauer P, Schmidt T, Schulte T, Riess O. Autosomal dominant cerebellar ataxias: clinical features, genetics, and pathogenesis. Lancet Neurol. 2004;3:291–304. doi: 10.1016/S1474-4422(04)00737-9. [DOI] [PubMed] [Google Scholar]
- 62.Cooper PE, Fernstrom MH, Rostad OP, Leeman SE, Martin JB. The regional distribution of somatostatin, substance P and neurotensin in human brain. Brain Res. 1981;218(1–2):219–232. doi: 10.1016/0006-8993(81)91302-0. [DOI] [PubMed] [Google Scholar]
- 63.Mai JK, Stephens PH, Hope A, Cuello AC. Substance P in the human brain. Neurosci. 1986;17(3):709–739. doi: 10.1016/0306-4522(86)90041-2. [DOI] [PubMed] [Google Scholar]
- 64.Cramer H, Rissler K, Rosler N, Strubel D, Schaudt D, Kuntzmann F. Immunoreactive substance P and somatostatin in the cerebrospinal fluid of senile parkinsonian patients. Eur Neurol. 1989;29(1):1–5. doi: 10.1159/000116366. [DOI] [PubMed] [Google Scholar]
- 65.Matsuishi T, Sakai T, Nagamitsu S, Shoji H, Ueda N, Kaneko S, Kano T, Iwashita H, Kato H. Decreased cerebrospinal fluid levels of substance P in Machado-Joseph disease. J Neurol Sci. 1996;142(1–2):107–110. doi: 10.1016/0022-510x(96)00159-1. [DOI] [PubMed] [Google Scholar]
- 66.Vink R, Donkin JJ, Cruz MI, Nimmo AJ, Cernak I. A substance P antagonist increases brain intracellular free magnesium concentration after diffuse traumatic brain injury in rats. J Am Coll Nutr. 2004;23(5):538S–540S. doi: 10.1080/07315724.2004.10719398. [DOI] [PubMed] [Google Scholar]
- 67.McIntosh TK, Smith DH, Meaney DF, Kotapka MJ, Gennarelli TA, Graham DI. Neuropathological sequelae of traumatic brain injury: relationship to neurochemical and biomechanical mechanisms. Lab Invest. 1996;74:315–342. [PubMed] [Google Scholar]
- 68.Lobato RD, Sarabia R, Cordobes F, Rivas JJ, Adrados A, Cabrera A, Gomez P, Madera A, Lamas E. Posttraumatic cerebral hemispheric swelling. Analysis of 55 cases studied with computerized tomography. J Neurosurg. 1988;68:417–423. doi: 10.3171/jns.1988.68.3.0417. [DOI] [PubMed] [Google Scholar]
- 69.Sarabia R, Lobato RD, Rivas JJ, Cordobes F, Rubio J, Cabrera A, Gomez P, Munoz MJ, Madera A. Cerebral hemisphere swelling in severe head injury patients. Acta Neurochir Suppl. 1988;42:40–46. doi: 10.1007/978-3-7091-8975-7_9. [DOI] [PubMed] [Google Scholar]
- 70.Woie K, Koller ME, Heyeraas KJ, Reed RK. Neurogenic inflammation in rat trachea is accompanied by increased negativity of interstitial fluid pressure. Circ Res. 1993;73:839–845. doi: 10.1161/01.res.73.5.839. [DOI] [PubMed] [Google Scholar]
- 71.Newbold P, Brain SD. An investigation into the mechanism of capsaicin-induced oedema in rabbit skin. Br J Pharmacol. 1995;114:570–577. doi: 10.1111/j.1476-5381.1995.tb17177.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Nimmo AJ, Cernak I, Heath DL, Hu X, Bennett CJ, Vink R. Neurogenic inflammation is associated with development of edema and functional deficits following traumatic brain injury in rats. Neuropeptides. 2004;38:40–47. doi: 10.1016/j.npep.2003.12.003. [DOI] [PubMed] [Google Scholar]
- 73.Malcangio M, Ramer MS, Jones MG, Mcmahon SB. Abnormal substance P release from the spinal cord following injury to primary sensory neurons. Eur J Neurosci. 2000;12(1):397–399. doi: 10.1046/j.1460-9568.2000.00946.x. [DOI] [PubMed] [Google Scholar]
- 74.Sharma HS, Nyberg F, Olsson Y, Dey PK. Alteration of substance P after trauma to the spinal cord: an experimental study in the rat. Neurosci. 1990;38(1):205–212. doi: 10.1016/0306-4522(90)90386-i. [DOI] [PubMed] [Google Scholar]
- 75.Donkin JJ, Turner RJ, Hassan I, Vink R. Substance P in traumatic brain injury. Prog Brain Res. 2007;161:97–109. doi: 10.1016/S0079-6123(06)61007-8. [DOI] [PubMed] [Google Scholar]
- 76.Yu Z, Cheng G, Huang X, Li K, Cao X. Neurokinin-1 receptor antagonist SR140333: a novel type of drug to treat cerebral ischemia. Neuroreport. 1997;8(9–10):2117–2119. doi: 10.1097/00001756-199707070-00006. [DOI] [PubMed] [Google Scholar]
- 77.Cadieux A, Springall DR, Mulderry PK, Rodrigo J, Chatei MA, Terenghi G, Bloom SR, Polak JM. Occurrence, distribution and ontogeny of CGRP immunoreactivity in the rat lower respiratory tract: effect of capsaicin treatment and surgical denervations. Neurosci. 1986;19(2):605–627. doi: 10.1016/0306-4522(86)90285-x. [DOI] [PubMed] [Google Scholar]
- 78.Kashiba H, Ueda Y, Senba E. Systemic capsaicin in the adult rat differentially affects gene expression for neuropeptides and neurotrophin receptors in primary sensory neurons. Neurosci. 1997;76(1):299–312. doi: 10.1016/s0306-4522(96)00334-x. [DOI] [PubMed] [Google Scholar]
- 79.Brookmeyer R, Gray S, Kawas C. Projections of Alzheimer’s disease in the United States and the public health impact of delaying disease onset. Am J Public Health. 1998;88(9):1337–1342. doi: 10.2105/ajph.88.9.1337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Maudsley S, Martin B, Luttrell LM. G protein-coupled receptor signaling complexity in neuronal tissue: implications for novel therapeutics. Curr Alzheimer Res. 2007;4(1):3–19. doi: 10.2174/156720507779939850. [DOI] [PubMed] [Google Scholar]
- 81.Querfurth HW, LaFerla FM. Alzheimer’s disease. N Engl J Med. 2010;362(4):329–344. doi: 10.1056/NEJMra0909142. [DOI] [PubMed] [Google Scholar]
- 82.Maudsley S, Mattson MP. Protein twists and turns in Alzheimer disease. Nat Med. 2006;12(4):392–393. doi: 10.1038/nm0406-392. [DOI] [PubMed] [Google Scholar]
- 83.Martin B, Brenneman R, Becker KG, Gucek M, Cole RN, Maudsley S. iTRAQ analysis of complex proteome alterations in 3×TgAD Alzheimer’s mice: understanding the interface between physiology and disease. PLoS One. 2008;3(7):e2750. doi: 10.1371/journal.pone.0002750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Kowall NW, Quigley BJ, Jr, Krause JE, Lu F, Kosofsky BE, Ferrante RJ. Substance P and substance P receptor histochemistry in human neurodegenerative diseases. Regul Pept. 1993;46(1–2):174–185. doi: 10.1016/0167-0115(93)90028-7. [DOI] [PubMed] [Google Scholar]
- 85.Barker R. Tachykinins, neurotrophism and neurodegenerative diseases: a critical review on the possible role of tachykinins in the aetiology of CNS diseases. Rev Neurosci. 1996;7(3):187–214. doi: 10.1515/revneuro.1996.7.3.187. [DOI] [PubMed] [Google Scholar]
- 86.Crystal HA, Davies P. Cortical substance P-like immunoreactivity in cases of Alzheimer’s disease and senile dementia of the Alzheimer type. J Neurochem. 1982;38(6):1781–1784. doi: 10.1111/j.1471-4159.1982.tb06665.x. [DOI] [PubMed] [Google Scholar]
- 87.Beal MF, Mazurek MF. Substance P-like immunoreactivity is reduced in Alzheimer’s disease cerebral cortex. Neurology. 1987;37:1205–1209. doi: 10.1212/wnl.37.7.1205. [DOI] [PubMed] [Google Scholar]
- 88.Quigley BJ, Jr, Kowall NW. Substance P-like immunoreactive neurons are depleted in Alzheimer’s disease cerebral cortex. Neuroscience. 1991;41(1):41–60. doi: 10.1016/0306-4522(91)90199-x. [DOI] [PubMed] [Google Scholar]
- 89.Yew DT, Li WP, Webb SE, Lai HWL, Zhang L. Neurotransmitters, peptides, and neural cell adhesion molecules in the cortices of normal elderly humans and Alzheimer patients: a comparison. Exp Geron. 1999;34(1):117–133. doi: 10.1016/s0531-5565(98)00017-5. [DOI] [PubMed] [Google Scholar]
- 90.Armstrong DM, Benzing WC, Evans J, Terry RD, Shields D, Hansen LA. Substance P and somatostatin coexist within neuritic plaques: implications for the pathogenesis of Alzheimer’s disease. Neuroscience. 1989;31:663–671. doi: 10.1016/0306-4522(89)90431-4. [DOI] [PubMed] [Google Scholar]
- 91.Pompei P, Severini R, Pediconi D, Angeletti M, Eleuteri A, Fattoretti P, Bertoni-Freddari C, Fioretti E. Regulation of preprotachykinin-A gene expression in an animal model of Alzheimer’s disease. J Histochem Cytochem. 2001;49(11):1469–1470. doi: 10.1177/002215540104901114. [DOI] [PubMed] [Google Scholar]
- 92.Willis M, Hutter-Paier B, Wietzorrek G, Windisch M, Humpel C, Knaus HG, Marksteiner J. Localization and expression of substance P in transgenic mice overexpressing human APP751 with the London (V717I) and Swedish (K670M/N671L) mutations. Brain Res. 2007;1143:199–207. doi: 10.1016/j.brainres.2007.01.080. [DOI] [PubMed] [Google Scholar]
- 93.Rösler N, Wichart I, Jellinger KA. Clinical significance of neurobiochemical profiles in the lumbar cerebrospinal fluid of Alzheimer’s disease patients. J Neural Transm. 2001;108(2):231–246. doi: 10.1007/s007020170091. [DOI] [PubMed] [Google Scholar]
- 94.Marksteiner J, Saria A, Krause JE. Comparative distribution of neurokinin B-, substance P-and enkephalin-like immunoreactivities and neurokinin B messenger RNA in the basal forebrain of the rat: evidence for neurochemical compartmentation. Neuroscience. 1992;51:107–120. doi: 10.1016/0306-4522(92)90475-h. [DOI] [PubMed] [Google Scholar]
- 95.Marksteiner J, Sperk G, Krause JE. Distribution of neurons expressing neurokinin B in the rat brain: immunohistochemistry and in situ hybridization. J Comp Neurol. 1992;317:341–356. doi: 10.1002/cne.903170403. [DOI] [PubMed] [Google Scholar]
- 96.Ahmed MM, Hoshino H, Chikuma T, Yamada M, Kato T. Effect of memantine on the levels of glial cells, neuropeptides, and peptide-degrading enzymes in rat brain regions of ibotenic acid-treated Alzheimer’s disease model. Neuroscience. 2004;126:639–649. doi: 10.1016/j.neuroscience.2004.04.024. [DOI] [PubMed] [Google Scholar]
- 97.Kart E, Jocham G, Muller CP, Schlomer C, Brandao ML, Huston JP, Souza Silva MA. Neurokinin-1 receptor antagonism by SR140333: enhanced in vivo ACh in the hippocampus and promnestic post-trial effects. Peptides. 2004;25:1959–1969. doi: 10.1016/j.peptides.2004.07.003. [DOI] [PubMed] [Google Scholar]
- 98.Souza Silva MA, Hasenohrl RU, Tomaz C, Schwarting RK, Huston JP. Differential modulation of frontal cortex acetylcholine by injection of substance P into the nucleus basalis magnocellularis region in the freely-moving vs. the anesthetized preparation. Synapse. 2000;38:243–253. doi: 10.1002/1098-2396(20001201)38:3<243::AID-SYN3>3.0.CO;2-G. [DOI] [PubMed] [Google Scholar]
- 99.Li SH, Li XJ. Huntingtin-protein interactions and the pathogenesis of Huntington’s disease. Trends Genet. 2004;20(3):146–154. doi: 10.1016/j.tig.2004.01.008. [DOI] [PubMed] [Google Scholar]
- 100.Martin B, Golden E, Keselman A, Stone M, Mattson MP, Egan JM, Maudsley S. Therapeutic perspectives for the treatment of Huntington’s disease: treating the whole body. Histol Histopathol. 2008;23(2):237–250. doi: 10.14670/hh-23.237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Landles C, Bates GP. Huntingtin and the molecular pathogenesis of Huntington’s disease. Fourth in molecular medicine review series. EMBO Rep. 2004;5(10):958–963. doi: 10.1038/sj.embor.7400250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Martin B, Golden E, Carlson OD, Pistell P, Zhou J, Kim W, Frank BP, Thomas S, Chadwick WA, Greig NH, Bates GP, Sathasivam K, Bernier M, Maudsley S, Mattson MP, Egan JM. Exendin-4 improves glycemic control, ameliorates brain and pancreatic pathologies, and extends survival in a mouse model of Huntington’s disease. Diabetes. 2009;58(2):318–328. doi: 10.2337/db08-0799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Martin JB. Huntington’s disease: new approaches to an old problem. The Robert Wartenberg lecture. Neurology. 1984;34(8):1059–1072. doi: 10.1212/wnl.34.8.1059. [DOI] [PubMed] [Google Scholar]
- 104.Ferrante RJ, Kowall NW, Richardson EP, Jr, Bird ED, Martin JB. Topography of enkephalin, substance P and acetylcholinesterase staining in Huntington’s disease striatum. Neurosci. 1986;71(3):283–288. doi: 10.1016/0304-3940(86)90634-8. [DOI] [PubMed] [Google Scholar]
- 105.Augood SJ, Faull RLM, Love DR, Emson PC. Reduction in enkephalin and substance P messenger RNA in the striatum of early grade Huntington’s disease: a detailed cellular in situ hybridization study. Neurosci. 1996;72(4):1023–1036. doi: 10.1016/0306-4522(95)00595-1. [DOI] [PubMed] [Google Scholar]
- 106.Richfield EK, Vonsattel JP, MacDonald ME, Sun Z, Deng YP, Reiner A. Selective loss of striatal preprotachykinin neurons in a phenocopy of Huntington’s disease. Mov Disord. 2002;17(2):327–332. doi: 10.1002/mds.10032. [DOI] [PubMed] [Google Scholar]
- 107.Graveland GA, Williams RS, DiFiglia M. Evidence for degenerative and regenerative changes in neostriatal spiny neurons in Huntington’s disease. Science. 1985;227(4688):770–773. doi: 10.1126/science.3155875. [DOI] [PubMed] [Google Scholar]
- 108.Park S, Yan Y, Satriotomo I, Vemuganti R, Dempsey RJ. Substance P is a promoter of adult neural progenitor cell proliferation under normal and ischemic conditions. J Neurosurg. 2007;107(3):593–599. doi: 10.3171/JNS-07/09/0593. [DOI] [PubMed] [Google Scholar]
- 109.Morcuende S, Gadd CA, Peters M, Moss A, Harris EA, Sheasby A, Fisher AS, De Filipe C, Mantyh PW, Rupniak NMJ, Giese KP, Hunt SP. Increased neurogenesis and brain-derived neurotrophic factor in neurokinin-1 receptor gene knockout mice. Euro J Neurosci. 2003;18(7):1828–1836. doi: 10.1046/j.1460-9568.2003.02911.x. [DOI] [PubMed] [Google Scholar]
- 110.McCutcheon JE, Fisher AS, Guzdar E, Wood SA, Lightman SL, Hunt SP. Genetic background influences the behavioural and molecular consequences of neurokinin-1 receptor knockout. Euro J Neurosci. 2008;27(3):683–690. doi: 10.1111/j.1460-9568.2008.06043.x. [DOI] [PubMed] [Google Scholar]
- 111.Chadwick W, Magnus T, Martin B, Keselman A, Mattson MP, Maudsley S. Targeting TNF-alpha receptors for neurotherapeutics. Trends Neurosci. 2008;31(10):504–511. doi: 10.1016/j.tins.2008.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Lieb K, Fiebich BL, Berger M, Bauer J, Schulze-Osthoff K. The neuropeptide substance P activates transcription factor NF-kappa B and kappa B-dependent gene expression in human astrocytoma cells. J Immunol. 1997;159(10):4952–4958. [PubMed] [Google Scholar]
- 113.Fiebich BL, Schleicher S, Butcher RD, Craig A, Lieb K. The neuropeptide substance P activates p38 mitogen-activated protein kinase resulting in IL-6 expression independently from NF-kappa B. J Immunol. 2000;165(10):5606–5611. doi: 10.4049/jimmunol.165.10.5606. [DOI] [PubMed] [Google Scholar]
- 114.Marriott I, Bost KL. IL-4 and IFN-gamma up-regulate substance P receptor expression in murine peritoneal macrophages. J Immunol. 2000;165(1):182–191. doi: 10.4049/jimmunol.165.1.182. [DOI] [PubMed] [Google Scholar]
- 115.Marriott I, Bost KL. Expression of authentic substance P receptors in murine and human dendritic cells. J Neuroimmunol. 2001;114(1–2):131–141. doi: 10.1016/s0165-5728(00)00466-5. [DOI] [PubMed] [Google Scholar]
- 116.Martin FC, Anton PA, Gornbein JA, Shanahan F, Merrill JE. Production of interleukin-1 by microglia in response to substance P: role for a non-classical NK-1 receptor. J Neuroimmunol. 1993;42(1):53–60. doi: 10.1016/0165-5728(93)90212-h. [DOI] [PubMed] [Google Scholar]
- 117.Kiefer R, Lindholm D, Kreutzberg GW. Interleukin-6 and transforming growth factor-beta 1 mRNAs are induced in rat facial nucleus following motoneuron axotomy. Eur J Neurosci. 1993;5(7):775–781. doi: 10.1111/j.1460-9568.1993.tb00929.x. [DOI] [PubMed] [Google Scholar]
- 118.Streit WJ, Hurley SD, McGraw TS, Semple-Rowland SL. Comparative evaluation of cytokine profiles and reactive gliosis supports a critical role for interleukin-6 in neuron-glia signaling during regeneration. J Neurosci Res. 2000;61(1):10–20. doi: 10.1002/1097-4547(20000701)61:1<10::AID-JNR2>3.0.CO;2-E. [DOI] [PubMed] [Google Scholar]
- 119.Kalra PS, Dube MG, Kalra SP. The effects of interleukin 1 beta on the hypothalamic tachykinin, neurokinin A. Brain Res. 1994;662(1–2):178–184. doi: 10.1016/0006-8993(94)90810-9. [DOI] [PubMed] [Google Scholar]
- 120.De Laurentiis A, Candolfi M, Pisera D, Seilicovich A. Effects of lipopolysaccharide on neurokinin A content and release in the hypothalamic-pituitary axis. Regul Pept. 2003;111(1–3):91–5. doi: 10.1016/s0167-0115(02)00258-6. [DOI] [PubMed] [Google Scholar]
- 121.McCluskey LP, Lampson LA. Local immune regulation in the central nervous system by substance P vs. glutamate. J Neuroimmunol. 2001;116(2):136–146. doi: 10.1016/s0165-5728(01)00295-8. [DOI] [PubMed] [Google Scholar]
- 122.Amadoro G, Pieri M, Ciotti MT, Carunchio I, Canu N, Calissano P, Zona C, Severini C. Substance P provides neuroprotection in cerebellar granule cells through Akt and MAPK/Erk activation: evidence for the involvement of the delayed rectifier potassium current. Neuropharma. 2007;52(6):1366–1377. doi: 10.1016/j.neuropharm.2007.01.020. [DOI] [PubMed] [Google Scholar]
- 123.Lallemend F, Lefebvre PP, Hans G, Rigo JM, Van de Water TR, Moonen G, Malgrange B. Substance P protects spiral ganglion neurons from apoptosis via PKC-Ca2+-MAPK/ERK pathways. J Neurochem. 2003;87(2):508–521. doi: 10.1046/j.1471-4159.2003.02014.x. [DOI] [PubMed] [Google Scholar]
- 124.Calvo N, Reiriz J, Pérez-Navarro E, Alberch J. Tachykinins protect cholinergic neurons from quinolinic acid excitotoxicity in striatal cultures. Brain Res. 1996;740(1–2):323–328. doi: 10.1016/s0006-8993(96)00879-7. [DOI] [PubMed] [Google Scholar]
- 125.Pieri M, Amadoro G, Carunchio I, Ciotti MT, Quaresima S, Florenzano F, Calissano P, Possenti R, Zona C, Severini C. SP protects cerebellar granule cells against beta-amyloid-induced apoptosis by down-regulation and reduced activity of Kv4 potassium channels. Neuropharmacology. 2010;58(1):268–276. doi: 10.1016/j.neuropharm.2009.06.029. [DOI] [PubMed] [Google Scholar]
- 126.Yankner BA, Duffy LK, Kirschner DA. Neurotrophic and neurotoxic effects of amyloid beta protein: reversal by tachykinin neuropeptides. Science. 1990;250(4978):279–282. doi: 10.1126/science.2218531. [DOI] [PubMed] [Google Scholar]
- 127.Kowall NW, Beal MF, Busciglio J, Duffy LK, Yanker BA. An in vivo model for the neurodegenerative effects of beta amyloid and protection by substance P. Proc Natl Acad Sci. 1991;88(16):7247–7251. doi: 10.1073/pnas.88.16.7247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Sankhala KK, Pandya DM, Sarantopoulos J, Soefje SA, Giles FJ, Chawla SP. Prevention of chemotherapy induced nausea and vomiting: a focus on aprepitant. Expert Opin Drug Metab Toxicol. 2009;5(12):1607–1614. doi: 10.1517/17425250903451675. [DOI] [PubMed] [Google Scholar]
- 129.Kramer MS, Cutler N, Feighner J, Shrivastava R, Carman J, Sramek JJ, Reines SA, Liu G, Snavely D, Wyatt-Knowles E, Hale JJ, Mills SG, MacCoss M, Swain CJ, Harrison T, Hill RG, Hefti F, Scolnick EM, Cascieri MA, Chicchi GG, Sadowski S, Williams AR, Hewson L, Smith D, Carlson EJ, Hargreaves RJ, Rupniak NMJ. Distinct mechanism for antidepressant activity by blockade of central substance P receptors. Science. 1998;281(5383):1640–1645. doi: 10.1126/science.281.5383.1640. [DOI] [PubMed] [Google Scholar]
- 130.Rupniak NMJ, Carlson EC, Harrison T, Oates B, Seward E, Owen S, de Felipe C, Hunt S, Wheeldon A. Pharmacological blockade or genetic deletion of substance P (NK(1)) receptors attenuates neonatal vocalisation in guinea-pigs and mice. Neuropharma. 2000;39(8):1413–1421. doi: 10.1016/s0028-3908(00)00052-6. [DOI] [PubMed] [Google Scholar]
- 131.Kramer MS, Winokur A, Kelsey J, Preskorn SH, Rothschild AJ, Snavely D, Ghosh K, Ball WA, Reines SA, Munjack D, Apter JT, Cunningham L, Kling M, Bari M, Getson A, Lee Y. Demonstration of the efficacy and safety of a novel substance P (NK1) receptor antagonist in major depression. Neuropsychopharmacology. 2004;29(2):385–392. doi: 10.1038/sj.npp.1300260. [DOI] [PubMed] [Google Scholar]
- 132.Van Belle S, Lichinitser MR, Navari RM, Garin AM, Decramer MLA, Riviere A, Thant M, Brestan E, Bui B, Eldridge K, De Smet M, Michiels N, Reinhardt RR, Carides AD, Evans JK, Gertz BJ. Prevention of cisplatin-induced acute and delayed emesis by the selective neurokinin-1 antagonists, L-758, 298 and MK-869. Cancer. 2002;94(11):3032–3041. doi: 10.1002/cncr.10516. [DOI] [PubMed] [Google Scholar]
- 133.Kramer JH, Mak IT, Phillips TM, Weglicki WB. Dietary magnesium intake influences circulating pro-inflammatory neuropeptide levels and loss of myocardial tolerance to postischemic stress. Exp Biol Med. 2003;228(6):665–673. doi: 10.1177/153537020322800604. [DOI] [PubMed] [Google Scholar]
- 134.Heath DL, Vink R. Traumatic brain axonal injury produces sustained decline in intracellular free magnesium concentration. Brain Res. 1996;738(1):150–153. doi: 10.1016/0006-8993(96)00957-2. [DOI] [PubMed] [Google Scholar]
- 135.Maudsley S, Martin B, Luttrell LM. The origins of diversity and specificity in G protein-coupled receptor signaling. J Pharmacol Exp Ther. 2005;314(2):485–494. doi: 10.1124/jpet.105.083121. [DOI] [PMC free article] [PubMed] [Google Scholar]