This book is copyright. Apart from any fair dealing for the purposes of private study, research, criticism or review as permitted under the Copyright Act, no part may be reproduced, stored in a retrieval system, or transmitted, in any form or by any means, electronic, mechanical, photocopying, recording or otherwise without the prior written permission. Address all inquiries to the Director at the above address.
NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.
Vink R, Nechifor M, editors. Magnesium in the Central Nervous System [Internet]. Adelaide (AU): University of Adelaide Press; 2011.
Abstract
Apoptosis is a distinctive feature in the physiology of the developing brain, but also a key event in pathological conditions of the adult brain. The basic mechanisms executing cell death by apoptosis are conserved among different tissues and in different conditions, while the initiating event(s) may be more specific. Magnesium appears to be an important player in the process, though it might exert opposite actions depending on extra/intracellular availability. Extracellular magnesium deficiency induces apoptosis, mainly through increased oxidative stress, while intracellular magnesium mobilization from intracellular stores and consequent increase of cytosolic free magnesium seem to act in the effector phase. The molecular mechanism and the physio-pathological meaning of these findings await further characterization. The issue is even more complex in the context of the brain, where many concurring factors may determine a pro- or anti-apoptotic environment. A deeper understanding of the yin-yang role of magnesium in apoptosis may cast light on the basic processes that regulate cell fate, and consequently may open up novel opportunities for a successful therapeutic intervention for all the pathological conditions where excessive and undue apoptosis takes place.
Cell death by apoptosis
Multicellular organisms maintain their homeo- stasis thanks to a tightly regulated mode of cell death. Programmed cell death, or apoptosis, is an internally controlled suicide program consisting in a stereotyped sequence of biochemical and morphological changes that allow the cell to die without adversely affecting its neighbours, i.e. without causing inflammation. As such, apoptosis is distinct from necrosis, the traumatic form of cell death whereby massive disruption of the surrounding tissue takes place. A cell may be doomed to die either by apoptosis or necrosis depending on the intensity and duration of the stimulus, the rapidity of the death process, and the extent of ATP depletion suffered by the cell.
Apoptosis occurs in numerous physiological, adaptive and pathological events: 1) during development; 2) as a homeostatic mechanism to maintain cell populations in proliferating tissues; 3) as a defence mechanism such as in immune reactions; 4) following cell injury from various agents; and 5) in aging. There is general agreement that this form of cell death is an energy-dependent cascade of molecular events leading to cell shrinkage with maintenance of organelle integrity; long-recognized features of apoptosis are protein cleavage and cross-linking, chromatin condensation, DNA fragmentation and, ultimately, phagocytic recognition and engulfment of the dying cell. The process of apoptosis is triggered by a diverse range of cell signals, which may originate either extracellularly (i.e. death receptor ligation with their cognate ligands, extrinsic pathway) or intracellularly (e.g. DNA damage or organelle dysfunction, intrinsic pathway).
The apoptotic cascade consists of three parts: first, cells process the receptor-mediated death signal (initiation phase, somewhat more cell- specific), then get ready to implement apoptosis (effector phase, more conserved among different tissues) and finally commit suicide (degradation phase). During the initiation phase, the receptor- mediated death signal activates an intracellular cascade of events including: 1) activation of initiator caspases (caspase-8, for example), that can act early in the cell death process before, or independently of, mitochondrial changes; 2) increase in levels of oxyradicals and Ca2+; 3) transcription and translocation of pro-apoptotic Bcl-2 family members (Bax and Bad) to the mitochondrial membrane. The execution phase involves: 1) increased mitochondrial Ca2+ and oxyradical levels; 2) opening of permeability transition pores (PTP) in the mitochondrial membrane; 3) release of cytochrome c and many other apoptosis-inducing factors from the mitochondrial matrix into the cytosol; 4) formation of a molecular complex (the apopto- some) between cytochrome c and apoptotic protease-activating factor 1 (Apaf-1), that recruits and activates pro-caspase-9. Activated caspase-9, in turn, activates caspase-3, which begins the degradation phase: proteins are cleaved by various caspases and packed by trans- glutaminases, while nucleic acids are cleaved by activated Ca2+/Mg2+ endonucleases, which results in the formation of apoptotic bodies and subsequent phagocytosis by elicited macro- phages or neighbouring cells. It is interesting to underline the role of mitochondria in triggering apoptosis. Whether or not a direct injury jeopardizing the efficiency of these energy producing organelles occurs, mitochondria play a pivotal role in the cell death decision, as apoptotic factors are released by active mechanisms, which can be unrelated to mitochondrial damage (Tait and Green, 2010).
Role of Magnesium in Apoptosis
In the attempt to review the role of magnesium in apoptosis, one should first of all keep in mind that the effects of extracellular magnesium availability may differ from those due to intracellular magnesium fluxes or mobilization. In fact, decreased extracellular magnesium availability does not necessarily reflect an intracellular depletion, as demonstrated by the fact that in vivo serum magnesium levels are not a reliable marker of pathological hypo- magnesemia. Sensitivity to extracellular magnesium availability is highly tissue-specific, and can be translated in molecular terms by considering the expression and activity of magnesium-specific cation channels, such as the transient receptor potential melastatin (TRPM) -6 and -7 channels, which, by modulating trans- membrane cation fluxes, regulate the intracellular ion concentration (Schlingmann et al., 2007). Furthermore, intracellular magnesium is tightly buffered to meet specific metabolic requirements. As discussed elsewhere in this book, large changes in total cell magnesium occur with little or no change in cytosolic free magnesium, suggesting that the changes in total magnesium are due to changes in bound or sequestered magnesium (Murphy, 2000). ATP is the most important intracellular magnesium buffer. Following massive ATP hydrolysis intra- cellular free magnesium transiently increases and is subsequently released into the extracellular environment. As a consequence, in a dying cell an increase in ionized magnesium occurs. Therefore, caution should be used also when investigating the role of intracellular magnesium as a player in the apoptotic program, in order to discriminate coincidental from causative events.
In conclusion, whether considering extracellular magnesium availability or intracellular magnesium concentration, the story is more complicated than it appears at first glance. We will attempt to discuss the issue by reviewing the latest available data in view of the complex cellular physiology of magnesium.
Extracellular magnesium and apoptosis
Several important aspects of magnesium biochemistry and physiology point to a possible role for this cation in the apoptotic process. As discussed in other chapters of this book, magnesium is a key modulator of cell proliferation and metabolism and, most importantly, magnesium availability appears to affect the occurrence of oxidative stress. Many studies are available in the literature on the subject, but the most convincing ones investigate the effects of hypomagnesemia in vivo. In these circumstances, however, other factors, including inflammation, cytokine production and activation of phagocyte oxidative burst, concur to what is referred to as the pro- oxidant effect of hypomagnesemia (Mazur et al., 2007). Induction of apoptosis mediated by oxidative stress following magnesium deprivation has been documented in several tissues. Accelerated thymus involution, a classical example of apoptosis, was found in magnesium- deficient rats (Malpuech-Brugère et al., 1999). Dietary magnesium deficiency also induced apoptosis in cardiovascular tissues (Altura et al., 2009; Tejero-Taldo et al., 2007) and in the liver (Martin et al., 2003; Martin et al., 2008). In addition to the induction of reactive oxygen species, low magnesium availability could in principle trigger apoptosis by affecting DNA structure. This occurs not only by promoting oxidative DNA damage, but also by impairing DNA repair mechanisms, since magnesium is required for several crucial DNA repair enzyme activities, e.g. endonucleases, ligases, topo- isomerases (Hartwig, 2001).
Intracellular magnesium and apoptosis
As to intracellular events, several in vitro studies have suggested a promoting role for magnesium in apoptosis. An early increase in intracellular magnesium seems to follow both extrinsic and intrinsic induction of apoptosis (Patel et al., 1994; Chien et al., 1999; Zhang et al., 2005). The most straightforward explanation is that the increase in intracellular magnesium concentration is necessary for stimulating the activity of Ca2+/ Mg2+dependent endonucleases, which perform the apoptotic event par excellence, i.e. nucleosomal DNA fragmentation. Interestingly, however, the source of intracellular magnesium was hypothesized to be in the mitochondria (Chien et al., 1999), which is particularly appealing, as it is well established that mitochondria play a central role in the onset of the apoptotic program. Other lines of evidence point to the same direction. While the opening of the permeability transition pores appears to be dispensable for the release of cytochrome c, this is not the case for the presence of magnesium, which instead seems an absolute requirement (Eskes et al., 1998; Kim et al., 2000). Another study (Salvi et al., 2004) indicated that an apoptotic compound, gliotoxin, can specifically activate a magnesium efflux system from mitochondria in conditions of preserved mitochondrial integrity (i.e. high membrane potential, no swelling and retention of other ions). Most importantly, it has been shown that mitochondria act in fact as magnesium stores, and that Mg2+ release can occur following Ca2+ release and prior to ATP hydrolysis (Kubota et al., 2005). It is noteworthy that the same Authors have recently demonstrated that glutamate administration to rat hippocampal neurons triggers the same pathway, whereby Ca2+ accumulation in the mitochondria is required for Mg2+ release from the organelles (Shindo et al., 2010). The presence of a specific mitochondrial channel for magnesium, Mrs2, (Kolisek et al., 2003) corroborates these findings. As the activity of Mrs2 is dependent from membrane potential, it can be speculated that stored magnesium might be released through the channel upon depolarization. Intriguingly, in a recent paper Mrs2 expression has been associated with resistance to drug-induced apoptosis in cancer cells (Chen et al., 2009): by upregulating magnesium uptake into mitochondria, Mrs2 might counteract the increase in cytosolic magnesium that seems to be necessary for the execution of the apoptotic program via the mitochondrial pathway (Wolf and Trapani, 2009). In conclusion, the latest findings seem to concur to suggest a finer involvement of magnesium in the apoptotic cascade: not just a biochemical factor, but rather a crucial control element in life vs death decisions (see Table 1 for a summary). At present, these are only attractive speculations; most importantly, it has to be clarified whether the increase in intracellular magnesium occurring following an apoptotic stimulus is just a coin- cidental event (for example, due to mitochondrial depolarization) or rather a causative determinant in the downstream signalling cascade.
Neuronal Apoptosis
In contrast to the rapid turnover of cells in proliferative/renewing tissues, neurons commonly survive for the entire lifetime of the organism — this enduring nature of neurons is necessary for maintaining the function of those cells within neuronal circuits. However, during development of the central and peripheral nervous systems, many neurons undergo apoptosis during a time window that coincides with the process of synaptogenesis (Oppenheim, 1991). Initial overproduction of neurons, followed by death of some, is probably an adaptive process that provides enough neurons to form nerve cell circuits that are precisely matched to their functional specifications. However, the persistence of neurons throughout life to preserve brain function implies that a considerable evolutionary pressure was placed on the development of mechanisms that guarded against neuronal death and/or promoted neuronal survival and plasticity.
Trophic factors
Like all cells, neuronal survival requires trophic support. Viktor Hamburger and Rita Levi- Montalcini described in a seminal paper that the survival of developing neurons is directly related to the availability of their innervating targets (Hamburger and Levi-Montalcini, 1949). This laid the foundation for the neurotrophin hypothesis, which proposed that immature neurons compete for target-derived trophic factors that are in limited supply. Neurotrophins, which include nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and neurotrophins 3 and 4/5 (Lewin and Barde, 1996), generally activate and ligate the Trk receptors (TrkA, TrkB and TrkC), which are cell-surface receptors with intrinsic tyrosine kinase activity. They can autophosphorylate; for instance, after the binding of NGF to TrkA, the receptor phosphorylates several tyrosine residues within its own cytoplasmic tail. These phosphotyrosines in turn serve as docking sites for other molecules such as phospholipase Cg, phosphoinositide 3- kinase (PI(3)K) and adaptor proteins such as Shc, and these signal transduction molecules coordinate neuronal survival through the Akt and MAP kinase signalling pathways (Hennigan et al., 2007). Several neurotrophic factors and cytokines use a survival pathway involving the transcription factor NF-κB (Mattson and Camandola, 2000).
The neurotrophin hypothesis predicts correctly that neuronal survival requires a positive survival signal. It does not, however, provide a concrete hypothesis as to how neurons die in the absence of trophic support; it was assumed that neurons die simply of passive starvation. In 1988, using cultured sympathetic neurons as a model system, Johnson and colleagues showed that inhibition of RNA and protein synthesis blocked sympathetic neuronal cell death induced by nerve growth factor (NGF) deprivation (Martin et al., 1988), providing the first tangible evidence that neurons might actually actively instigate their own demise.
Glutamate receptors
Lack of neurotrophic support is undeniably the best-studied signal that may trigger apoptosis during development of the nervous system. However, most neurons in the mammalian central nervous system possess receptors for another trigger of apoptosis, glutamate.
Glutamate is a major excitatory neurotransmitter with a crucial role in neural development, synaptic plasticity, and learning and memory under physiological conditions (Riedel et al., 2003). Glutamate receptors are classified into several classes. In this context, we will go into some detail only for N-methyl-D-aspartate (NMDA) receptors. NMDA receptors are tetra- heteromeric ligand-gated ion channels that open upon the binding of glutamate. Magnesium binds the channel pore in a voltage-dependent manner; thus, at normal physiological resting membrane potential, the NMDA receptor is blocked by Mg2+. Synaptic release of glutamate causes Na+ influx through α-amino-3-hydroxy-5-methyl-4-isoxazole- propionate (AMPA) receptors in the postsynaptic cell, resulting in partial membrane depolarization sufficient to lift the Mg2+ block and activate the channel. The activated NMDA receptor is permeable to Na+ but, crucially, also to Ca2+. This Ca2+ influx mediates most of the physiological effects of NMDA receptor activity, leading to postsynaptic depolarization and action potential in the postsynaptic neuron.
Physiological levels of synaptic NMDA receptor activity are essential for neuronal survival (Hetman and Kharebava, 2006). However, regulation of glutamatergic neurotransmission is critical, as improper management of glutamate levels and glutamate receptor activity may impair not only its signalling properties, but can lead to cell death. The concept of excitotoxicity was first proposed by John Olney in 1969 as a toxic effect of excessive or prolonged activation of receptors by excitatory amino acids (Olney, 1969). Although the molecular pathways involved in excitotoxicity are still not fully understood at the present, there is a wealth of evidence suggesting that over- stimulation of glutamate receptors produces multiple adverse effects including impairment of intracellular calcium homeostasis, dysfunction of mitochondria and endoplasmic reticulum, increase in nitric oxide (NO) production and free radicals, persistent activation of proteases and kinases, increases in expression of pro-death transcription factors and immediate early genes (IEGs), ultimately leading to apoptosis (Wang and Qin, 2010). Excitotoxicity might mediate neuronal damage in various neurological disorders including ischemia and traumatic brain injury (Arundine and Tymianski, 2004) and neurodegen- erative diseases (Lipton and Rosenberg, 1994; Rego and Oliveira, 2003); it has also been implicated in neonatal brain injury (Johnston, 2005). NMDA receptors are important mediators of glutamate- induced excitotoxicity, as calcium entering through over-activated NMDA receptors results in more cell death as opposed to calcium entering through non-NMDA glutamate receptors or voltage-gated calcium channels (Cristofanilli and Akopian, 2006), which maybe due in part to their high permeability to Ca2+ and incomplete desensitization.
Thus, responses to NMDA receptor activity follow a classical hormetic dose-response curve: both too much and too little can be harmful (Hardingham, 2009).
Pathological stimuli
Another trigger of neuronal death is increased oxidative stress, whereby free radicals (such as the superoxide anion radical and the hydroxyl radical) damage cellular lipids, proteins and nucleic acids by disrupting chemical bonds in those molecules. Metabolic stress, in which levels of glucose, oxygen and other molecules required for ATP (energy) production are decreased, and environmental toxins may also initiate neuronal apoptosis (Mattson, 2000). These stimuli are mostly involved in aging and pathological conditions, such as acute or chronic neuro- degenerative diseases. The classical and most studied pathway of oxidation-induced apoptosis consists in cell-damage triggered phosphorylation of p53 and transcription of pro-apoptotic factors like Bid or Bad leading to mitochondrial- dependent activation of caspase 9.
The genetic and environmental factors that trigger neuronal apoptosis may be different in various physiological and pathological settings and can vary from those active in other tissues, but most of the subsequent biochemical events that execute the cell death process are highly conserved and shared with all other cell types. Thus, the key components of the apoptosis program in neurons, like that of other cell types, are Apaf-1 and proteins in the Bcl-2 and caspase families. Nevertheless, different types of neurons, and neurons at different developmental stages, express different combinations of Bcl-2 and caspase family members, which is one way of providing the specificity of regulation (Yuan and Yankner, 2000).
Tissue specific factors
An alternative way to control cell death or survival is through the expression of tissue- specific proteins that affect signal transduction reactions and may be either pro-apoptotic or pro- survival.
The prostate apoptosis response-4 (Par-4) protein was identified as being upregulated in prostate tumour cells undergoing apoptosis, but is now known to be essential in developmental and pathological neuronal death (Guo et al., 1998). Levels of Par-4 increase rapidly in response to various apoptotic stimuli through enhanced translation of Par-4 messenger RNA. Par-4 acts at an early stage of the apoptotic cascade prior to caspase activation and mitochondrial dys- function, by a mechanism that may involve inhibition of the antiapoptotic transcription factor NF-kβ and suppression of Bcl-2 expression and/or function (Mattson et al., 1999).
Rai is an Shc-related adaptor protein whose expression is exclusively restricted to the nervous system. It exerts a prosurvival function in neuronal cells by activating the PI3K/Akt signalling pathway (Pelicci et al., 2002).
A neuron-specific splice variant of the variable subunit Bβ was characterized, which is induced upon neuronal differentiation, targets protein phosphatase 2A to mitochondria and accelerates neuronal cell apoptosis after survival factor deprivation (Dagda et al., 2003).
Lately, a brain-specific isoform of mitochondrial apoptosis-inducing factor, AIF2, has been isolated (Hangen et al., 2010). AIF2 dimerizes with AIF1 and has a stronger membrane anchorage. Therefore it is conceivable the neuron-specific AIF2 may have been ‘designed’ to be retained in mitochondria and to minimize its potential neurotoxic activity.
In conclusion, although mature neurons are among the most long-lived cell types in mammals, immature neurons die in large numbers and the regulation of apoptosis has a major role in sculpting the developing brain. Furthermore, not only is apoptosis important in regulating neuronal development, but it might also be a cardinal feature in the adult brain in several pathological conditions that will be detailed in the section on neurodegenerative diseases.
Role of Magnesium in Neuronal Apoptosis
Deciphering the role of magnesium in the apoptotic process is further complicated in the nervous system, as the blood brain barrier may restrain serum magnesium availability within the CNS. Indeed, this has been advocated as the cause for the failure of clinical trials assessing magnesium therapy in acute and chronic brain injury (Vink et al., 2009).
A significant part of the literature on the effects of magnesium on apoptosis in the brain is related to perinatal brain, as magnesium is often used as a treatment for pre-eclampsia/eclampsia and preterm labour. The basis for its use as a tocolytic is due to its observed effects in reducing myometrial contractility through extra- and intracellular mechanisms of action (Fomin et al., 2006). However, the use of magnesium sulphate as a drug in obstetrical medicine for both mother and fetus is highly controversial. A recent meta- analysis of all trials has recommended the use of magnesium for neuroprotection in the preterm fetus, as it was both safe and effective (Doyle et al., 2009). Nevertheless, some caution is mandatory, as several reports point to detrimental effects of magnesium for the fetal brain, depending on the dose (Mittendorf et al., 2006; Dribben et al., 2009), on the period of neurodevelopment when the exposure occurred (Dribben et al., 2009) or on the level of stress experienced (Krueger et al., 2001). In addition, some papers report a pro-apoptotic action of magnesium in placental tissues (Black et al., 2001; Gude et al., 2000). The major hurdle to establishing protective or deleterious effects of tocolytic magnesium to the developing brain is that magnesium’s effects on preterm infant’s brains, and the mechanisms of those effects, are not understood.
As discussed earlier, neuronal apoptosis can be triggered by three main mechanisms: 1) lack of growth factors; 2) overstimulation of glutamate receptors; and 3) oxidative stress. Magnesium could play a (different) role in each of these signalling pathways.
First, most growth factors work through receptor tyrosine kinases (RTKs) which require two magnesium ions for maximal activity. Kinases downstream of these RTKs, such as PI -kinase and the anti-apoptotic kinase Akt require magnesium both for their activation and activity. Therefore, extracellular magnesium could modulate the effect of growth factors on CNS cells, or activate their second messenger systems directly, ultimately affecting viability, proliferation, and apoptosis in these cells.
Secondly, under conditions of normal membrane polarization magnesium is known to block the NMDA glutamate receptor and prevent ion flow through the channel. Therefore, in principle, magnesium should protect against excitotoxicity- induced apoptosis. Nevertheless, as already discussed, normal physiological patterns of NMDA receptor activity promote neuroprotection against both apoptotic and excitotoxic insults; vice versa, NMDA receptor blockade can promote neuronal death outright or render neurons vulnerable to secondary trauma (Hardingham, 2009). Indeed, most NMDA antagonists have failed miserably as neuroprotective agents in clinical trials, in large part because of intolerable side effects (Lipton, 2006). Moreover, it has been shown that various drugs that inhibit neuronal activity, including NMDA antagonists, trigger widespread apoptotic neurodegeneration in the developing brain and cause long-term neurobehavioral deficits; in the infant rodent brain, peak sensitivity to this effect is in the P3 to P7 period (Ikonomidou et al., 1999). Thus, although other mechanisms may play a role, it seems likely that the neuroapoptogenic action demonstrated for magnesium in some studies could be mediated by its action at NMDA receptors.
Third, as mentioned earlier, magnesium deprivation may induce or exacerbate oxidative stress, another trigger of neuronal death, especially in pathological conditions. In this regard, magnesium supplementation might well have a neuroprotective action.
Apoptosis in neurodegenerative diseases
As already mentioned, neuronal death by apoptosis is a distinctive physiological feature of the developing brain; once synaptogenesis is over, the remaining neurons will usually survive throughout life to preserve brain function. Unfortunately, however, many people experience excessive death of specific neuronal sub- populations as a result of chronic diseases including Alzheimer’s disease, Parkinson’s disease, Huntington’s disease and amyotrophic lateral sclerosis. Neuronal death may also occur as a consequence of acute conditions, such as stroke or traumatic brain and spinal cord injury. For each of these disorders, apoptosis has been implicated as the main form of cell death, though it is very difficult to demonstrate it for both technical and ethical reasons. Much of the evidence supporting an apoptotic mode of neuronal death comes from studies of animal and cell-culture models of neurodegenerative disorders (Mattson, 2000).
Physiological apoptosis in the developing brain and pathological apoptosis in the adult brain share similar molecular mechanisms in the effector phase, but may differ in the initiation. Whereas trophic factor withdrawal has a prominent role in apoptosis during development, toxic insults resulting from biochemical or genetic accidents seem to be the triggering event in neurodegenerative disorders. An emerging theme is the toxicity of abnormal protein structures or aggregates (Yuan and Yankner, 2000). Indeed, a defining feature of Alzheimer’s disease is accumulation of amyloid plaques formed by aggregates of amyloid-β peptide (Aβ). Parkinson’s disease is characterized by the appearance of α-synuclein oligomers. Huntington’s disease is caused by expansions of a trinucleotide (CAG) sequence in the huntingtin gene producing a protein containing increased polyglutamine repeats. The neuropathological signature of amyotrophic lateral sclerosis (ALS) is the presence of ubiquitinated inclusions immuno- reactive for the proteins TDP-43 and/or FUS/TLS in the cytoplasm of motor neurons. Moreover, mutations in specific genes may predispose to neuronal degeneration, e.g. presenilin in Alzheimer's disease, parkin in Parkinson's disease, huntingtin in Huntington's disease, and Cu/Zn- superoxide dismutase (SOD) in ALS (please refer to the dedicated chapters in this book for further details).
The identification of specific genetic and environmental factors responsible for chronic neurodegenerative disorders has bolstered evidence for a shared pathway of neuronal apoptosis involving oxidative stress, perturbed calcium homeostasis and mitochondrial dysfunction ultimately converging on caspase activation (Zündorf and Reiser, 2010; Gibson et al., 2010).
In Alzheimer's disease, toxic forms of Aβ may induce Ca2+ influx into neurons by formation of an oligomeric pore in the plasma membrane, thereby rendering neurons vulnerable to excitotoxicity and apoptosis (Supnet and Bezprozvanny, 2010). Aβ also generates hydrogen peroxide and hydroxyl radicals, producing membrane lipid peroxidation and consequently membrane depolarization and excitotoxicity through NMDA receptor channels and voltage- gated Ca2+ channels (Sultana and Butterfield, 2010; Butterfield et al., 2010). In addition, Aβ accumulates in mitochondria, impairs the activity of complexes III and IV of the respiratory chain, causes elevated cytoplasmic Ca2+ levels and oxidative stress, and reduces ATP synthesis, further increasing Ca2+ overload and oxidative stress (Moreira et al., 2010).
Mitochondrial dysfunction is a defect occurring early in the pathogenesis of both sporadic and familial Parkinson's disease. Mitochondrial association of α-synuclein in cells was linked to impairment of respiratory complex I activity, oxidative modification of mitochondrial proteins, and increased levels of Ca2+ and nitric oxide (Navarro and Boveris, 2009).
Mitochondrial Ca2+ overload is also a decisive commitment step for Huntington's disease. Mutant huntingtin facilitates opening of the PTP and directly impairs the mitochondrial function. Furthermore, huntingtin forms a ternary complex with other proteins, which causes Ca2+ release from the ER and renders neurons more sensitive to Ca2+-mediated cellular dysfunction (Damiano et al., 2010).
The mutations in SOD responsible for ALS do not decrease antioxidant activity of the enzyme, but result in the gain of an adverse pro-apoptotic activity that may involve increased peroxidase activity. Through interactions with hydrogen peroxide or superoxide anion, the mutant enzyme may induce oxidative damage to membranes and disturbances in mitochondrial function that make neurons vulnerable to excitotoxic apoptosis (Vucic and Kiernan, 2009). Also, stroke and trauma, and the consequent brain ischemia, initiate biochemical and molecular events involving many of the same neurodegenerative cascades that occur in the chronic neurodegenerative diseases described above, namely excitotoxicity, calcium overload and oxidative stress (Arundine and Tymianski, 2004).
In conclusion, despite the concurrence of diverse genetic and environmental factors for acute and chronic neurodegenerative disorders, such conditions possess a common denominator as to the ultimate mechanisms executing neuronal death, which are identical to those involved in normal brain development and all converge to mitochondrial dysfunction. On one hand, these findings testify the evolutionary importance of conserving fundamental processes. On the other hand, they call for further research to understand the basic mechanisms and the key regulators of neuronal fate, as novel opportunities for targeted therapeutic intervention may arise.
Role of magnesium in neurodegenerative diseases
As detailed above, there is increasing evidence that neuronal death by apoptosis is associated with both acute and chronic neurodegenerative disorders. Interestingly, brain free magnesium levels have been shown to decline in a number of such pathologies (Vink et al., 2009). As a consequence, a considerable research effort has been directed toward establishing the mechanisms of such decline and the potential for magnesium administration as a neuroprotective agent.
Brain magnesium decline is a ubiquitous feature of traumatic brain injury and is associated with the development of motor and cognitive deficits. Experimentally, parenteral administration of magnesium up to 12 h post-trauma restores brain magnesium homeostasis and profoundly improves both motor and cognitive outcome. Although the mechanism of action is unclear, magnesium has been shown to attenuate a variety of secondary injury factors, including brain edema, cerebral vasospasms, glutamate excitotoxicity, calcium-mediated events, lipid per- oxidation, mitochondrial permeability transition, and apoptosis. Disappointingly, magnesium therapy has failed in clinical trials. Increase in brain free magnesium concentration seems to be essential to confer neuroprotection, and intravenous magnesium administration only marginally increases CSF magnesium concentration, which suggests that the integrity of the blood- brain barrier and the regulation of magnesium in the cerebrospinal fluid are largely maintained following acute brain injury and limit magnesium bioavailability in the brain (Vink and Nimmo, 2009).
Magnesium therapy has also been described in the clinical stroke literature, with similar negative results, though magnesium was shown to be beneficial in a subgroup of patients with noncortical, or lacunar, strokes (Ginsberg, 2008). Magnesium may protect via multiple mechanisms, including NMDA receptor blockade, inhibition of excitatory neurotransmitter release, blockade of calcium channels, as well as vascular smooth muscle relaxation, and may be more bioavailable, as it is well known that the blood brain barrier around infarcted tissue is highly permeable, thus potentially facilitating local magnesium entry to the injured tissues.
Intriguingly, the cation channel TRPM7, which is crucial for Mg2+ homeostasis and cell survival (Schmitz et al., 2003), seems to be a critical mediator of anoxic cell death. It appears that TRPM7 gating lies downstream of NMDA receptor-mediated NO free-radical production. In this paradigm, excitotoxicity is likely the initiating signal in a cell death cascade that involves nNOS activation with subsequent peroxynitrite production, which in turn activates TRPM7 channels. Calcium influx through TRPM7 then creates a positive feedback loop of ROS production, which eventually kills the cell (Aarts and Tymianski, 2005). TRPM7 suppression made neurons resistant to ischemic death after brain ischemia and preserved neuronal morphology and function; also, it prevented ischemia-induced deficits in long-term potentiation and preserved performance in memory tasks (Sun et al., 2009). The role of the magnesium-specific channel TRPM6 in these processes is currently unclear, but, given its localization in the brain, its permeability to Mg2+ and its functional interaction with TRPM7, it may be involved in neuronal death as well (Cook et al., 2009). At present, the role of TRPM6/7 channels in magnesium transport seems less important than their facilitation of other cation fluxes, but it cannot be ruled out, especially considering the known involvement of magnesium in excitotoxicity and oxidative stress, as discussed earlier.
With respect to chronic brain degeneration, the first connection between magnesium and three apparently dissimilar neurodegenerative dis- orders (ALS, Parkinson's and Alzheimer's diseases) came from the observation that those conditions occurred at extraordinarily high rates in geographically separate foci among three genetically different, homogenous western Pacific populations (Durlach et al., 1997). Intensive research conducted over the years led to the identification of two candidate environmental factors: 1) severely low levels of Ca2+ and Mg2+ in the soil and drinking water coupled with abnormal mineral metabolism; and 2) the putative neurotoxin β-methylamino-L- alanine (L-BMAA), derived from the cycad plant, a traditional food source in Guam. Experimental findings supported this hypothesis: animals fed diets that mimic the mineral composition in the disease foci environment showed signs of neuronal damage; of the various combinations of Ca2+ and Mg2+ contents tested, exposure to low Mg2+ (one-fifth of the normal level) was more deleterious, causing significant loss of dopaminergic neurons in the substantia nigra (Oyanagi et al., 2006). Furthermore, motor neurons are selectively more vulnerable to L- BMAA toxicity, and these toxic effects are mediated through [Ca2+]i rises via Ca-A/K channels and other pathways, and subsequent reactive oxygen species generation (Rao et al., 2006).
In the hyperendemic foci in the Western Pacific, the unique mineral composition of the environment creates a condition of increased oxidative stress because low Mg2+ levels cause lipid peroxidation while transition metals act as redox catalysts causing increased ROS production. Therefore, the genes whose function is most likely to be affected are ion channels expressed in the CNS and modulated by oxidative stress. TRPM7 and TRPM2 fulfil these conditions; their functional properties are interconnected with calcium and magnesium homeostasis, oxidative stress, mitochondrial dysfunction, and immune mechanisms, all principal suspects in neurodegeneration (Hermosura and Garruto, 2007). Indeed, variants of both genes have been found in Guamanian neurodegenerative disorders and may contribute to their pathogenesis (Hermosura et al., 2008; Hermosura et al., 2005).
Despite the evidence implicating magnesium in the aetiology of neurodegenerative diseases, the therapeutic effects of magnesium supplementation are not well documented. Magnesium administration proved beneficial in an in vitro model of Parkinson's disease, where in particular magnesium concentration seems critical in the onset of the disease (Hashimoto et al., 2008), but there is still much to be done to fully appreciate the therapeutic potential of magnesium.
Conclusion
An extensive and up-to-date review of the literature shows that drawing a clear-cut conclusion on the role of magnesium in neuroapoptosis is not an easy task. Magnesium may play pleiotropic roles in different stages of the apoptotic process, that range from modulating growth factor- and Ca2+-dependent signal transduction to affecting oxidative stress- related reactions. From this point of view, increased extracellular magnesium availability should in principle be considered anti- apoptogenic. On the other hand, an increase in cytosolic free magnesium, which seems to be released from mitochondrial stores, appears to accompany the intrinsic apoptotic process. The pathophysiological meaning of this occurrence in the context of apoptosis induction/execution awaits to be defined (see Figure 1 for a summary).
In a more physiological basis, magnesium availability in the brain should be considered within a more complex and interconnected picture, where other conditions, such as developmental stage, inflammation, acute or chronic injuries, blood brain barrier integrity, and so on, may concur in determining a pro or anti- apoptotic environment.
We believe that further investigation into the role of magnesium in the molecular mechanisms of apoptosis is warranted for a deeper understanding of the basic processes that regulate cell fate, and ultimately for a successful therapeutic intervention when these processes go awry.
Acknowledgments
Supported by the Italian Ministry of Education, University and Research PRIN 2007ZT39FN and Linea D1 2004-2009.
References
- Aarts MM, Tymianski M. TRPM7 and ischemic CNS injury. Neuroscientist. 2005;11:116–23. [PubMed: 15746380]
- Altura BM, Shah NC, Jiang XC, Li Z, Perez-Albela JL, Sica AC, Altura BT. Short-term magnesium deficiency results in decreased levels of serum sphingomyelin, lipid peroxidation, and apoptosis in cardiovascular tissues. Am J Physiol Heart Circ Physiol. 2009;297:H86–92. [PubMed: 19429813]
- Arundine M, Tymianski M. Molecular mechanisms of glutamate-dependent neurodegeneration in ischemia and traumatic brain injury. Cell Mol Life Sci. 2004;61:657–68. [PubMed: 15052409]
- Black S, Yu H, Lee J, Sachchithananthan M, Medcalf RL. Physiologic concentrations of magnesium and placental apoptosis: prevention by antioxidants. Obstet Gynecol. 2001;98:319–24. [PubMed: 11506852]
- Butterfield DA, Bader Lange ML, Sultana R. Involvements of the lipid peroxidation product, HNE, in the pathogenesis and progression of Alzheimer's disease. Biochim Biophys Acta. 2010;1801(8):924–9. [PMC free article: PMC2886168] [PubMed: 20176130]
- Chen Y, Wei X, Yan P, Han Y, Sun S, Wu K, Fan D. Human mitochondrial Mrs2 protein promotes multidrug resistance in gastric cancer cells by regulating p27, cyclin D1 expression and cytochrome C release. Cancer Biol Ther. 2009;8:607–14. [PubMed: 19242098]
- Chien MM, Zahradka KE, Newell MK, Freed JH. Fas-induced B cell apoptosis requires an increase in free cytosolic magnesium as an early event. J Biol Chem. 1999;274:7059–66. [PubMed: 10066762]
- Cook NL, van den Heuvel C, Vink R. Are the transient receptor potential melastatin (TRPM) channels important in magnesium homeostasis following traumatic brain injury? Magnes Res. 2009;22:225–34. [PubMed: 20228000]
- Cristofanilli M, Akopian A. Calcium channel and glutamate receptor activities regulate actin organization in salamander retinal neurons. J Physiol. 2006;575:543–54. [PMC free article: PMC1819466] [PubMed: 16777935]
- Dagda RK, Zaucha JA, Wadzinski BE, Strack S. A developmentally regulated, neuron-specific splice variant of the variable subunit Bbeta targets protein phosphatase 2A to mitochondria and modulates apoptosis. J Biol Chem. 2003;278:24976–85. [PubMed: 12716901]
- Damiano M, Galvan L, Déglon N, Brouillet E. Mitochondria in Huntington's disease. Biochim Biophys Acta. 2010;1802:52–61. [PubMed: 19682570]
- Deshmukh M, Johnson EM. Programmed cell death in neurons: focus on the pathway of nerve growth factor deprivation-induced death of sympathetic neurons. Mol Pharmacol. 1997;51:897–906. [PubMed: 9187255]
- Doyle LW, Crowther CA, Middleton P, Marret S, Rouse D. Magnesium sulphate for women at risk of preterm birth for neuroprotection of the fetus. Cochrane Database Syst Rev. 2009;(1):CD004661. [PubMed: 19160238]
- Durlach J, Bac P, Durlach V, Durlach A, Bara M. Are age-related neurodegenerative diseases linked with various types of magnesium depletion? Magnes Res. 1997;10:339–53. Guiet- Bara A. [PubMed: 9513930]
- Eskes R, Antonsson B, Osen-Sand A, Montessuit S, Richter C, Sadoul R, Mazzei G, Nichols A, Martinou JC. Bax-induced cytochrome C release from mitochondria is independent of the permeability transition pore but highly dependent on Mg2+ ions. J Cell Biol. 1998;143:217–24. [PMC free article: PMC2132823] [PubMed: 9763433]
- Fomin VP, Gibbs SG, Vanam R, Morimiya A, Hurd WW. Effect of magnesium sulfate on contractile force and intracellular calcium concentration in pregnant human myometrium. Am J Obstet Gynecol. 2006;194:1384–90. [PubMed: 16647924]
- Gibson GE, Starkov A, Blass JP, Ratan RR, Beal MF. Cause and consequence: mitochondrial dysfunction initiates and propagates neuronal dysfunction, neuronal death and behavioral abnormalities in age-associated neurodegenerative diseases. Biochim Biophys Acta. 2010;1802:122–34. [PMC free article: PMC2790547] [PubMed: 19715758]
- Ginsberg MD. Neuroprotection for ischemic stroke: past, present and future. Neuropharmacology. 2008;55:363–89. [PMC free article: PMC2631228] [PubMed: 18308347]
- Gude NM, Stevenson JL, Moses EK, King RG. Magnesium regulates hypoxia-stimulated apoptosis in the human placenta. Clin Sci (Lond). 2000;98:375–80. [PubMed: 10731470]
- Guo Q, Fu W, Xie J, Luo H, Sells SF, Geddes JW, Bondada V, Rangnekar VM, Mattson MP. Par-4 is a mediator of neuronal degeneration associated with the pathogenesis of Alzheimer disease. Nature Med. 1998;4:957–62. [PubMed: 9701251]
- Hamburger V, Levi-Montalcini R. Proliferation, differentiation and degeneration in the spinal ganglia of the chick embryo under normal and experimental conditions. J Exp Zool. 1949;111:457–502. [PubMed: 18142378]
- Hangen E, De Zio D, Bordi M, Zhu C, Dessen P, Caffin F, Lachkar S, Perfettini JL, Lazar V, Benard J, Fimia GM, Piacentini M, Harper F, Pierron G, Vicencio JM, Bénit P, de Andrade A, Höglinger G, Culmsee C, Rustin P, Blomgren K, Cecconi F, Kroemer G, Modjtahedi N. A brain-specific isoform of mitochondrial apoptosis-inducing factor: AIF2. Cell Death Differ. 2010;17:1155–66. [PubMed: 20111043]
- Hardingham GE. Coupling of the NMDA receptor to neuroprotective and neurodestructive events. Biochem Soc Trans. 2009;37(Pt 6):1147–60. [PMC free article: PMC2837198] [PubMed: 19909238]
- Hartwig A. Role of magnesium in genomic stability. Mutat Res. 2001;475:113–21. [PubMed: 11295157]
- Hashimoto T, Nishi K, Nagasao J, Tsuji S, Oyanagi K. Magnesium exerts both preventive and ameliorating effects in an in vitro rat Parkinson disease model involving 1-methyl-4-phenylpyridinium (MPP+) toxicity in dopaminergic neurons. Brain Res. 2008;1197:143–51. [PubMed: 18242592]
- Hennigan A, O'Callaghan RM, Kelly AM. Neurotrophins and their receptors: roles in plasticity, neurodegeneration and neuroprotection. Biochem Soc Trans. 2007;35(Pt 2):424–7. [PubMed: 17371291]
- Hermosura MC, Cui AM, Go RC, Davenport B, Shetler CM, Heizer JW, Schmitz C, Mocz G, Garruto RM, Perraud AL. Altered functional properties of a TRPM2 variant in Guamanian ALS and PD. Proc Natl Acad Sci USA. 2008;105:18029–34. [PMC free article: PMC2584682] [PubMed: 19004782]
- Hermosura MC, Garruto RM. TRPM7 and TRPM2-Candidate susceptibility genes for Western Pacific ALS and PD? Biochim Biophys Acta. 2007;1772:822–35. [PMC free article: PMC1994999] [PubMed: 17395433]
- Hermosura MC, Nayakanti H, Dorovkov MV, Calderon FR, Ryazanov AG, Haymer DS, Garruto RM. A TRPM7 variant shows altered sensitivity to magnesium that may contribute to the pathogenesis of two Guamanian neurodegenerative disorders. Proc Natl Acad Sci USA. 2005;102:11510–5. [PMC free article: PMC1183597] [PubMed: 16051700]
- Hetman M, Kharebava G. Survival signaling pathways activated by NMDA receptors. Curr Top Med Chem. 2006;6:787–99. [PubMed: 16719817]
- Ikonomidou C, Bosch F, Miksa M, Bittigau P, Vockler J, Dikranian K, Tenkova TI, Stefovska V, Turski L, Olney JW. Blockade of NMDA receptors and apoptotic neurodegeneration in the developing brain. Science. 1999;283:70–74. [PubMed: 9872743]
- Johnston MV. Excitotoxicity in perinatal brain injury. Brain Pathol. 2005;15:234–40. [PMC free article: PMC8095755] [PubMed: 16196390]
- Kim TH, Zhao Y, Barber MJ, Kuharsky DK, Yin XM. Bid-induced cytochrome c release is mediated by a pathway independent of mitochondrial permeability transition pore and Bax. J Biol Chem. 2000;275:39474–81. [PubMed: 10982793]
- Kolisek M, Zsurka G, Samaj J, Weghuber J, Schweyen RJ, Schweigel M. Mrs2p is an essential component of the major electrophoretic Mg2+ influx system in mitochondria. EMBO J. 2003;22:1235–44. [PMC free article: PMC151051] [PubMed: 12628916]
- Krueger RC Jr, Santore MT, Dawson G, Schwartz NB. Increased extracellular magnesium modulates proliferation in fetal neural cells in culture. Brain Res Dev Brain Res. 2001;127:99–109. [PubMed: 11334997]
- Kubota T, Shindo Y, Tokuno K, Komatsu H, Ogawa H, Kudo S, Kitamura Y, Suzuki K, Oka K. Mitochondria are intracellular magnesium stores: investigation by simultaneous fluorescent imagings in PC12 cells. Biochim Biophys Acta. 2005;1744:19–28. [PubMed: 15878394]
- Lewin GR, Barde YA. Physiology of the neurotrophins. Annu Rev Neurosci. 1996;19:289–317. [PubMed: 8833445]
- Lipton SA. Paradigm shift in neuroprotection by NMDA receptor blockade: memantine and beyond. Nat Rev Drug Discov. 2006;5:160–70. [PubMed: 16424917]
- Lipton SA, Rosenberg PA. Excitatory amino acids as a final common pathway for neurologic disorders. N Engl J Med. 1994;330:613–22. [PubMed: 7905600]
- Malpuech-Brugère C, Nowacki W, Gueux E, Kuryszko J, Rock E, Rayssiguier Y, Mazur A. Accelerated thymus involution in magnesium-deficient rats is related to enhanced apoptosis and sensitivity to oxidative stress. Br J Nutr. 1999;81:405–11. [PubMed: 10615213]
- Martin DP, Schmidt RE, Di Stefano PS, Lowry OH, Carter JG, Johnson EM Jr. Inhibitors of protein synthesis and RNA synthesis prevent neuronal death caused by nerve growth factor deprivation. J Cell Biol. 1988;106:829–844. [PMC free article: PMC2115082] [PubMed: 2450099]
- Martin H, Richert L, Berthelot A. Magnesium deficiency induces apoptosis in primary cultures of rat hepatocytes. J Nutr. 2003;133:2505–11. [PubMed: 12888628]
- Martin H, Uring-Lambert B, Adrian M, Lahlou A, Bonet A, Demougeot C, Devaux S, Laurant P, Richert L, Berthelot A. Effects of long-term dietary intake of magnesium on oxidative stress, apoptosis and ageing in rat liver. Magnes Res. 2008;21:124–30. [PubMed: 18705541]
- Mattson MP. Apoptosis in neurodegenerative disorders. Nat Rev Cell Mol Biol. 2000;1:120–9. [PubMed: 11253364]
- Mattson MP, Camandola S. NF-κB in neurodegenerative disorders. J Clin Invest. 2000;61:134–9. [PubMed: 10878585]
- Mattson MP, Duan W, Chan SL, Camandola S. Par-4: an emerging pivotal player in neuronal apoptosis and neurodegenerative disorders. J Mol Neurosci. 1999;13:17–30. [PubMed: 10691289]
- Mazur A, Maier JA, Rock E, Gueux E, Nowacki W, Rayssiguier Y. Magnesium and the inflammatory response: potential physiopathological implications. Arch Biochem Biophys. 2007;458:48–56. [PubMed: 16712775]
- Mittendorf R, Dammann O, Lee KS. Brain lesions in newborns exposed to high-dose magnesium sulfate during preterm labor. J Perinatol. 2006;26:57–63. [PubMed: 16319938]
- Moreira PI, Carvalho C, Zhu X, Smith MA, Perry G. Mitochondrial dysfunction is a trigger of Alzheimer's disease pathophysiology. Biochim Biophys Acta. 2010;1802:2–10. [PubMed: 19853658]
- Murphy E. Mysteries of magnesium homeostasis. Circ Res. 2000;86:245–8. [PubMed: 10679471]
- Olney JW. Brain lesions, obesity, and other disturbances in mice treated with monosodium glutamate. Science. 1969;164:719–721. [PubMed: 5778021]
- Oppenheim RW. Cell death during development of the nervous system. Annu Rev Neurosci. 1991;14:453–501. [PubMed: 2031577]
- Oyanagi K, Kawakami E, Kikuchi-Horie K, Ohara K, Ogata K, Takahama S, Wada M, Kihira T, Yasui M. Magnesium deficiency over generations in rats with special references to the pathogenesis of the Parkinsonism-dementia complex and amyotrophic lateral sclerosis of Guam. Neuropathology. 2006;26:115–28. [PubMed: 16708544]
- Patel T, Bronk SF, Gores GJ. Increases of intracellular magnesium promote glycodeoxycholate- induced apoptosis in rat hepatocytes. J Clin Invest. 1994;94:2183–92. [PMC free article: PMC330043] [PubMed: 7989573]
- Pelicci G, Troglio F, Bodini A, Melillo RM, Pettirossi V, Coda L, De Giuseppe A, Santoro M, Pelicci PG. The neuron-specific Rai (ShcC) adaptor protein inhibits apoptosis by coupling Ret to the phosphatidylinositol 3-kinase/Akt signaling pathway. Mol Cell Biol. 2002;22:7351–63. [PMC free article: PMC139827] [PubMed: 12242309]
- Rao SD, Banack SA, Cox PA, Weiss JH. BMAA selectively injures motor neurons via AMPA/kainate receptor activation. Exp Neurol. 2006;201:244–52. [PubMed: 16764863]
- Rego AC, Oliveira CR. Mitochondrial dysfunction and reactive oxygen species in excitotoxicity and apoptosis: implications for the pathogenesis of neurodegenerative diseases. Neurochem Res. 2003;28:1563–74. [PubMed: 14570402]
- Riedel G, Platt B, Micheau J. Glutamate receptor function in learning and memory. Behav Brain Res. 2003;140:1–47. [PubMed: 12644276]
- Salvi M, Bozac A, Toninello A. Gliotoxin induces Mg2+ efflux from intact brain mitochondria. Neurochem Int. 2004;45:759–64. [PubMed: 15234120]
- Schlingmann KP, Waldegger S, Konrad M, Chubanov V, Gudermann T. TRPM6 and TRPM7--Gatekeepers of human magnesium metabolism. Biochim Biophys Acta. 2007;1772:813–21. [PubMed: 17481860]
- Schmitz C, Perraud AL, Johnson CO, Inabe K, Smith MK, Penner R, Kurosaki T, Fleig A, Scharenberg AM. Regulation of vertebrate cellular Mg2+ homeostasis by TRPM7. Cell. 2003;114:191–200. [PubMed: 12887921]
- Shindo Y, Fujimoto A, Hotta K, Suzuki K, Oka K. Glutamate-induced calcium increase mediates magnesium release from mitochondria in rat hippocampal neurons. J Neurosci Res. 2010;88:3125–32. [PubMed: 20740499]
- Sultana R, Butterfield DA. Role of oxidative stress in the progression of Alzheimer's disease. J Alzheimers Dis. 2010;19:341–53. [PubMed: 20061649]
- Sun HS, Jackson MF, Martin LJ, Jansen K, Teves L, Cui H, Kiyonaka S, Mori Y, Jones M, Forder JP, Golde TE, Orser BA, Macdonald JF, Tymianski M. Suppression of hippocampal TRPM7 protein prevents delayed neuronal death in brain ischemia. Nat Neurosci. 2009;12:1300–7. [PubMed: 19734892]
- Supnet C, Bezprozvanny I. The dysregulation of intracellular calcium in Alzheimer disease. Cell Calcium. 2010;47:183–9. [PMC free article: PMC2834825] [PubMed: 20080301]
- Tait SW, Green DR. Mitochondria and cell death: outer membrane permeabilization and beyond. Nat Rev Mol Cell Biol. 2010;11:621–32. [PubMed: 20683470]
- Tejero-Taldo MI, Chmielinska JJ, Weglicki WB. Chronic dietary Mg2+ deficiency induces cardiac apoptosis in the rat heart. Magnes Res. 2007;20:208–12. [PubMed: 17972464]
- Vink R, Cook NL, van den Heuvel C. Magnesium in acute and chronic brain injury: an update. Magnes Res. 2009;22:158S–62S. [PubMed: 19780402]
- Vink R, Nimmo AJ. Multifunctional drugs for head injury. Neurotherapeutics. 2009;6:28–42. [PMC free article: PMC5084254] [PubMed: 19110197]
- Vucic S, Kiernan MC. Pathophysiology of neurodegeneration in familial amyotrophic lateral sclerosis. Curr Mol Med. 2009;9:255–72. [PubMed: 19355908]
- Wang Y, Qin ZH. Molecular and cellular mechanisms of excitotoxic neuronal death. Apoptosis. 2010;15:1382–402. [PubMed: 20213199]
- Wolf FI, Trapani V. Multidrug resistance phenotypes and MRS2 mitochondrial magnesium channel: two players from one stemness? Cancer Biol Ther. 2009;8:615–7. [PubMed: 19270501]
- Yuan J, Yankner BA. Apoptosis in the nervous system. Nature. 2000;407:802–9. [PubMed: 11048732]
- Zhang XY, Bai DC, Wu YJ, Li WG, Liu NF. Proanthocyanidin from grape seeds enhances anti- tumor effect of doxorubicin both in vitro and in vivo. Pharmazie. 2005;60:533–8. [PubMed: 16076082]
- Zhang XY, Li WG, Wu YJ, Bai DC, Liu NF. Proanthocyanidin from grape seeds enhances doxorubicin-induced antitumor effect and reverses drug resistance in doxorubicin-resistant K562/DOX cells. Can J Physiol Pharmacol. 2005;83:309–18. [PubMed: 15870845]
- Zündorf G, Reiser G. Calcium dysregulation and homeostasis of neural calcium in the molecular mechanisms of neurodegenerative diseases provide multiple targets for neuroprotection. Antioxid Redox Signa. 2010;14:1275–88. [PMC free article: PMC3122891] [PubMed: 20615073]
- Deficiency of calcium and magnesium induces apoptosis via scavenger receptor BI.[Life Sci. 2011]Deficiency of calcium and magnesium induces apoptosis via scavenger receptor BI.Feng H, Guo L, Gao H, Li XA. Life Sci. 2011 Mar 28; 88(13-14):606-12. Epub 2011 Feb 1.
- Review Preclinical development of novel anti-glioma drugs targeting the endoplasmic reticulum stress response.[Curr Pharm Des. 2011]Review Preclinical development of novel anti-glioma drugs targeting the endoplasmic reticulum stress response.Schönthal AH, Chen TC, Hofman FM, Louie SG, Petasis NA. Curr Pharm Des. 2011; 17(23):2428-38.
- End-to-End syndrome differentiation of Yin deficiency and Yang deficiency in traditional Chinese medicine.[Comput Methods Programs Biomed...]End-to-End syndrome differentiation of Yin deficiency and Yang deficiency in traditional Chinese medicine.Hu Q, Yu T, Li J, Yu Q, Zhu L, Gu Y. Comput Methods Programs Biomed. 2019 Jun; 174:9-15. Epub 2018 Oct 16.
- Review Cell (patho)physiology of magnesium.[Clin Sci (Lond). 2008]Review Cell (patho)physiology of magnesium.Wolf FI, Trapani V. Clin Sci (Lond). 2008 Jan; 114(1):27-35.
- Spatial Transcriptomic Analysis Using R-Based Computational Machine Learning Reveals the Genetic Profile of Yang or Yin Deficiency Syndrome in Chinese Medicine Theory.[Evid Based Complement Alternat...]Spatial Transcriptomic Analysis Using R-Based Computational Machine Learning Reveals the Genetic Profile of Yang or Yin Deficiency Syndrome in Chinese Medicine Theory.Zhang C, Tam CW, Tang G, Chen Y, Wang N, Feng Y. Evid Based Complement Alternat Med. 2022; 2022:5503181. Epub 2022 Mar 16.
- Magnesium and the Yin-Yang interplay in apoptosis - Magnesium in the Central Ner...Magnesium and the Yin-Yang interplay in apoptosis - Magnesium in the Central Nervous System
Your browsing activity is empty.
Activity recording is turned off.
See more...