There is an important unmet need to further improve the outcome of neonatal encephalopathy in term infants. Meta-analyses of large controlled trials now suggest that maternal magnesium sulfate (MgSO4) therapy is associated with a reduced risk of cerebral palsy and gross motor dysfunction after premature birth, but that it has no effect on death or disability. Because of this inconsistency, it remains controversial whether MgSO4 is clinically neuroprotective and, thus, it is unclear whether it would be appropriate to test MgSO4 for treatment of encephalopathy in term infants. We therefore systematically reviewed the preclinical evidence for neuroprotection with MgSO4 before or after hypoxic-ischemic encephalopathy (HIE) in term-equivalent perinatal and adult animals. The outcomes were highly inconsistent between studies. Although there were differences in dose and timing of administration, there was evidence that beneficial effects of MgSO4 were associated with confounding mild hypothermia and, strikingly, the studies that included rigorous maintenance of environmental temperature or body temperature consistently suggested a lack of effect. On balance, these preclinical studies suggest that peripherally administered MgSO4 is unlikely to be neuroprotective. Rigorous testing in translational animal models of perinatal HIE is needed before MgSO4 should be considered in clinical trials for encephalopathy in term infants.

Hypoxic-ischemic encephalopathy (HIE) around term remains a major cause of neonatal mortality and morbidity with lifelong chronic disabilities. Although therapeutic hypothermia for neonatal HIE improves survival without disability, with current protocols, nearly half of infants still die or survive with disability [1]. Thus, there is a crucial need to identify adjunct therapies to reduce the incidence and severity of HIE in near-term/term infants [2]. There has been considerable interest in magnesium sulfate (MgSO4) because magnesium alleviates excitotoxic damage in vitro by binding to the magnesium site on the NMDA (N-methyl-D-aspartate) glutamate channel [3]; there is some evidence that it may also reduce secondary inflammation and associated injury [4], stabilize cell membranes [5], inhibit free radical production [5] and improve cardiovascular stability [6].

Supporting this, there is now evidence from meta-analyses of randomized controlled trials that antenatal administration of MgSO4 is associated with a small but significant reduction in the risk of cerebral palsy and gross motor dysfunction in early childhood after preterm birth [7,8]. These meta-analyses included women in labor or at high risk of labor at ≤33 weeks gestation that received loading doses of 4-6 g of MgSO4 i.v. and/or a maintenance dose of 1-3 g/h or 5 g every 4 h i.m. However, although there was no significant change in the risk of death, there was a small trend in some of the trials, such that there was no net effect on overall death and disability [9,10]. Thus, there has been controversy whether these findings truly demonstrate a direct neuroprotective effect, and the exact mechanisms how MgSO4 may confer neurological benefit remain controversial. Further, there is no strong clinical evidence that MgSO4 is neuroprotective for near-term/term infants with HIE after maternal [11] or neonatal administration [12,13,14,15].

The purpose of this review was to systematically evaluate preclinical studies on magnesium for perinatal neuroprotection and identify knowledge gaps that need to be addressed before magnesium-induced neuroprotection could be considered for pragmatic trials at term.

In this review, an international group of researchers focusing on perinatal neuroprotection have examined the preclinical evidence for the therapeutic potential of MgSO4 for HIE. The studies were grouped into those that demonstrated improved or unaffected neural outcomes based on histological and/or behavioral assessments. Studies were further subdivided by species, age (perinatal vs. adult), type of insult used to induce HIE, treatment regime, timing of initiation of treatment (before or after the insult), extent of temperature monitoring (brain, core, body surface, ambient and none), main study outcomes (histological and/or behavioral) and survival time after treatment.

Studies were searched for on PubMed and MEDLINE (OvidSP) using the following terms: (brain injury OR hypoxia-ischemia OR cerebral ischemia) AND (magnesium OR MgSO4). Other sources used to identify studies included relevant original manuscripts and reviews. We did not set a date limit to the search. Studies were deemed eligible if they presented clear histological and/or behavioral outcomes from in vivo experiments that investigated magnesium for neuroprotection. Abstracts were initially identified and screened by an unbiased investigator from our study group and duplicated by another investigator. Selection of relevant studies was by group consensus.

Inclusion criteria for the perinatal studies were in vivo studies of rodents on postnatal day 6 (P6) to P21, sheep at 0.85 gestation and term piglets, as these developmental ages broadly reflect cortical maturity of term infants. We excluded in vitro studies and those that did not meet these age criteria. For the adult studies, inclusion criteria were in vivo studies of ischemic brain disease, i.e. global ischemia or permanent and temporary focal ischemia, as these are well-established experimental paradigms of cardiac arrest and stroke/subarachnoid hemorrhage with distinct and well-defined patterns of regional brain injury. We excluded studies that did not assess the effects of magnesium independent of adjuvant therapy.

The methodological quality of the studies was assessed according to whether temperature was controlled, treatment was randomized and investigators were blinded to the intervention during histological and behavioral assessments. The results are summarized in tables 1 and 2.

Table 1

Perinatal studies of magnesium for HIE in at-term or near-term equivalents

Perinatal studies of magnesium for HIE in at-term or near-term equivalents
Perinatal studies of magnesium for HIE in at-term or near-term equivalents
Table 2

Adult studies of magnesium for HI injury

Adult studies of magnesium for HI injury
Adult studies of magnesium for HI injury

Perinatal Studies of Magnesium for Term HIE

We identified and screened a total of 66 studies. Thirty-eight were excluded due to inappropriate developmental age, ex vivo studies or lack of assessment of the effects of magnesium independent of adjuvant therapy (adult studies only). Thus, 28 studies were surveyed in this study. Studies were then stratified by treatment regime, i.e. treatment before or after the insult, outcome and combination with other therapies.

Most of the neonatal and adult rodent studies measured infarct area or volume and performed semiquantitative assessments of neuronal apoptosis to examine neuronal loss/survival/severity of the insult. One neonatal rat study combined semiquantitative (graded) assessment of neuronal loss with the incidence of cyst formation and one study measured cerebral hemisphere weight as a broad indicator for improved neurodevelopmental outcome. In large animal studies, assessments of seizures in fetal sheep using electroencephalography and brain energy metabolism in piglets using magnetic resonance spectroscopy were combined with histopathological assessments of brain injury. The majority of the studies surveyed presented data as continuous variables, however, 5 perinatal studies presented data using graded assessment.

Fifteen studies examined the effectiveness of MgSO4 for neuroprotection in term or near-term HIE. The great majority of the studies (12/15) were performed in neonatal rats on P7 or P21, other studies used near-term fetal sheep (0.85 gestation) or 2- to 4-day-old neonatal piglets. In rats, cortical myelination is broadly comparable with 34 weeks of gestation in human infants on P7, and to late infancy by P21 [16]. In the 0.85 gestation sheep and term neonatal piglet, cortical maturity is broadly comparable with term human infants [17].

Most of the rodent studies used the Rice-Vannucci model of unilateral carotid artery ligation followed by a period of moderate hypoxia [16]. This functional paradigm of perinatal brain injury is associated with infarction of the middle cerebral artery region ipsilateral to the ligated carotid artery, with no overt cell death in the contralateral cerebral hemisphere. In sheep, cerebral injury was caused by either partial umbilical cord occlusion or repeated complete umbilical cord occlusion, leading to either a basal ganglion-predominant or a watershed pattern, as previously reviewed [18]. Similar regional brain injury has been observed in neonatal piglets following HIE induced by cerebral hypoperfusion with hypoxia [19].

Regime and Timing of Magnesium Delivery

The administration regimes for magnesium were highly variable. Magnesium was administered in single or multiple doses that ranged from 72 to 500 mg/kg. Typically, it was given by intraperitoneal bolus in neonatal rodent studies and continuous intravenous infusions to the fetus or neonate in large animal studies. Most post-insult studies initiated magnesium treatment within 1 h after insult.

Neurological Outcome and Survival Time after Near-Term or Term HIE

Seven (47%) of the perinatal studies surveyed reported improved neural outcomes with magnesium treatment alone (table 1) [20,21,22,23,24,25,26]. In 3 studies, MgSO4 was given before the start of HI [20,21,22]. In 4/7 studies, treatment was begun immediately after HI [24,25] or within 1.5 h [23,26]. One additional study reported improved histopathological outcome when MgSO4 was combined with oxygen-derived free radical scavengers, however, the effects of MgSO4 alone were not assessed [27]. Most of these studies used relatively short survival times of <1-5 days (4/9) or 7-13 days (4/9); just 1 study examined outcomes after 35 days of recovery [25]; this study was also the only one to report improved sensorimotor function.

Seven perinatal studies reported no improvement with MgSO4 after HI, all after short to medium survival times (2-11 days) [20,21,24,28,29,30,31,32]. All of the rodent studies and 1 piglet study showing no benefit involved treatment after the insult [21,24,28,29,31,32]. In the study in near-term fetal lambs, MgSO4 was infused from 2 h before umbilical cord occlusion and similarly showed no benefit [30].

In rodent studies, 2/7 that reported neuroprotection controlled the environmental temperature during HI [20,23], compared with 3/5 that found no effect [20,28,29]. None of the rodent studies directly measured brain or body temperature. The piglet study reported that rectal temperature was controlled [31,32]. Although details were not provided in the fetal sheep study [30], other studies have shown no significant effect of ischemia or asphyxia or drug treatments on cortical temperatures of fetal sheep [33,34].

Adult Studies of Magnesium for Cerebral Ischemia

All of the adult studies in rodent models of cerebral ischemia used either (1) global ischemia induced by bilateral carotid artery occlusion or (2) focal ischemia using middle cerebral artery occlusion. Eight studies reported improved histological outcome for MgSO4 alone [35,36,37,38,39,40,41,42], of which only 2/8 documented temperature at limited times after ischemia [35,41]. Seven studies reported no improvement in histological outcome with MgSO4 or MgCl2[43,44,45,46,47,48]. In all of these studies, postischemic temperature was monitored continuously and normothermia was maintained. Finally, 3 studies reported improved histological outcome when MgSO4 was combined with hypothermia, all of which included maintenance of the target temperatures [45,46,47]. Survival times varied from 1 to 7 days. No study in either group reported long-term outcome, and only 2 reported any behavioral outcome [38,47].

This systematic review shows that the effect of MgSO4 treatment before or shortly after acute hypoxia-ischemia (HI) at term or near-term equivalent was highly inconsistent between studies. Of considerable concern, none of the perinatal rodent studies suggesting benefit directly controlled brain or body temperature, and most did not adequately control environmental temperatures. Similarly, none of the studies suggesting benefit in adult rodents closely monitored core or brain temperature, and only 2 studies included limited/intermittent measurements after ischemia. Conversely, it is striking that the majority of studies that did control environmental or body temperature did not find significant protection. Many studies in adult rodents reported that beneficial effects of MgSO4 were associated with confounding mild hypothermia and that maintenance of normothermia abolished neuroprotection. Supporting this concern, large animal, ‘translational models' of perinatal injury in which body temperature was either directly controlled or known to be highly stable suggested lack of effect after 2 or 3 days of recovery [30,31,32].

Bioavailability

One of the great unknowns associated with MgSO4 for perinatal neuroprotection is whether direct or maternal administration can raise brain and cerebrospinal fluid magnesium concentrations to the level required for neuroprotection in the fetus or neonate. In culture, high concentrations of MgSO4 are required to reduce NMDA channel activity [3]. In this setting, increased extracellular magnesium is associated with recovery of high-energy phosphate stores, improved rates of protein synthesis and neuronal preservation in ischemic hippocampal neurons from fetal and adult rodents [49,50] and after global ischemia in adults when magnesium was given by direct intrahippocampal injection [51]. However, benefit required a 2- to 4-fold increase in extracellular magnesium concentrations. It is not clear whether it is possible to achieve such concentrations after peripheral administration. While in vivo studies have used measures of brain and cerebrospinal fluid excitotoxins, namely glutamate, to indirectly measure the local effect of systemically administered MgSO4, these measurements are relatively crude, have produced conflicting data and are confounded by a lack of body temperature monitoring [24,52]. Brain and cerebrospinal fluid magnesium concentrations are tightly controlled, such that in healthy dogs and rats, hypermagnesemia (3-4 times increased) was associated with modest changes in cerebrospinal fluid and tissue magnesium concentrations [53,54]. Prolonged systemic magnesium administration was associated with a much smaller increase in magnesium levels in brain parenchyma (from approximately 0.30 to 0.47 mmol/l) than the increase in the systemic circulation (from approximately 0.8 to 2.5 mmol/l) [55].

Vascular Effects

Alternatively, it is plausible that MgSO4 neuroprotection in preclinical studies may reflect vasodilation of cerebral microvessels through binding to calcium receptors in vascular smooth muscle [56]. In goats, both intravenously and after direct injection into the cerebral circulation, MgSO4 was associated with increased cerebral perfusion and reduced vascular resistance [57]. Thus, it is possible that pretreatment with MgSO4 in models of carotid ligation may promote an essentially artifactual improvement in perfusion during hypoxia [36,39,45].

Further, magnesium promotes vasodilation of peripheral vascular beds, including the mesenteric, renal, skeletal muscle and skin in rodents and larger animals [59]. Increased cutaneous perfusion is likely to increase heat loss through radiation; this may be considerable in small animals with a relatively large surface area and low thermal stability [60]. None of the perinatal studies assessed brain temperature; this may be important since in small animals, with relatively flat skulls, brain temperature can vary independently of body temperature [60]. Given that a mild reduction in body temperature can be neuroprotective [18,60] and that the majority of studies suggesting neuroprotection did not rigorously control brain or body temperature (tables 1, 2), it is plausible that magnesium may be associated with iatrogenic neuroprotection through hypothermia. Consistent with this, adult rodents treated with magnesium and allowed to self-regulate body temperature after global cerebral ischemia had mildly lower core temperatures and better hippocampal neuronal survival than controls [44]. Conversely, maintenance of normothermia after ischemia abolished the apparent neuroprotection.

Potential for Adjunct Therapy

No studies of combined hypothermia and magnesium therapy in perinatal animals were identified. In adult rats, pretreatment with MgSO4 before global cerebral ischemia [44], combined with mild spontaneous hypothermia after ischemia, reduced hippocampal neuronal loss more than MgSO4 or hypothermia alone [45]. Further, combined treatment with MgSO4 and mild hypothermia after permanent middle cerebral artery occlusion showed additive protection when administration of MgSO4 was delayed by 2 or 4 h [46]. Similarly, intracarotid infusion of cold MgSO4 (33-34°C) after focal brain ischemia was associated with a reduction in infarct volume, cerebral edema and neurological severity scores compared to saline or normothermic MgSO4 (37°C) infusion alone [47]. Thus, combination therapy is promising but it has not yet been tested for neonatal HIE. One trial of hypothermia with MgSO4 (Mag Cool) is currently listed on ClinicalTrials.Gov.

Survival Time after Treatment

The other major limitation of the perinatal studies identified in this review is that many used relatively short survival times, from 2.5 h to 5 days, and only 1 study reported improved neurobehavioral outcome. These shorter survival times provide important information about acute histological outcomes, however, brain injury can evolve for many weeks [61], and neurodevelopmental outcome can be discordant from histological outcomes [60]. In adult rodents treated with MgSO4 for cerebral injury, studies of survival times ≥7 days have rarely reported significant neuroprotection [44,45,48,62], and 9% of the adult studies surveyed in this review showed neuroprotective effects of magnesium per se after 7 days. Similarly, 43% of perinatal studies identified in the present review demonstrated no improvement in neurodevelopmental outcome after survival times of 7-11 days.

While we were not able to specifically assess publication bias, most of the studies randomized treatment and or used blinded assessors for histological and behavioral examinations. However, 2 perinatal studies (1 from each category) and 3 adult studies (2 showed improved outcomes and 1 showed no improvement) did not document randomization or blinding as part of their experimental protocol. Further, based on the major methodological differences in studies surveyed and lack of a common, specific outcome measure, we could not perform a meta-analysis of the studies surveyed. We found that all rodent studies that associated magnesium with neuroprotection did not perform adequate brain and/or body temperature monitoring, allowing for neuroprotection to be associated with confounding mild hypothermia rather than magnesium per se. Collectively, these methodological flaws indicate that further studies are required before attempting a meta-analysis of preclinical data for the neuroprotective benefit of magnesium.

This review of preclinical studies of MgSO4 for near-term HIE highlights the highly inconsistent histopathological impact of treatment both before and after the insult. These inconsistencies are likely related to a lack of temperature control during and after HI, along with variability in the dose, timing of treatment and survival time after the insult. These findings strongly suggest that clinical trials of MgSO4 for encephalopathy at term would be premature. Finally, despite very promising results from studies of global or focal ischemia in adult rodents, we were unable to identify any studies of MgSO4 combination therapy in the developing brain. Taken collectively, these observations suggest a crucial need for further testing in translational animal models of HIE before magnesium should be considered for pragmatic trials as a potential adjunct therapy with hypothermia.

The study was supported by the Health Research Council of New Zealand and the Auckland Medical Research Foundation.

The authors have no conflicts of interest to disclose. The study sponsors had no role in (1) the study design; (2) the collection, analysis and interpretation of the data; (3) the writing of the report, and (4) the decision to submit the paper for publication. No honorarium, grant or other form of payment was given to anyone to produce the paper.

1.
Edwards AD, Brocklehurst P, Gunn AJ, Halliday H, Juszczak E, Levene M, Strohm B, Thoresen M, Whitelaw A, Azzopardi D: Neurological outcomes at 18 months of age after moderate hypothermia for perinatal hypoxic ischaemic encephalopathy: synthesis and meta-analysis of trial data. BMJ 2010;340:c363.
2.
Robertson NJ, Tan S, Groenendaal F, van Bel F, Juul SE, Bennet L, Derrick M, Back SA, Valdez RC, Northington F, Gunn AJ, Mallard C: Which neuroprotective agents are ready for bench to bedside translation in the newborn infant? J Pediatr 2012;160:544.e4-552.e4.
3.
Zeevalk GD, Nicklas WJ: Evidence that the loss of the voltage-dependent Mg2+ block at the N-methyl-D-aspartate receptor underlies receptor activation during inhibition of neuronal metabolism. J Neurochem 1992;59:1211-1220.
4.
Sugimoto J, Romani AM, Valentin-Torres AM, Luciano AA, Ramirez Kitchen CM, Funderburg N, Mesiano S, Bernstein HB: Magnesium decreases inflammatory cytokine production: a novel innate immunomodulatory mechanism. J Immunol 2012;188:6338-6346.
5.
Hoffman DJ, Marro PJ, McGowan JE, Mishra OP, Delivoria-Papadopoulos M: Protective effect of MgSO4 infusion on NMDA receptor binding characteristics during cerebral cortical hypoxia in the newborn piglet. Brain Res 1994;644:144-149.
6.
Shokry M, Elsedfy GO, Bassiouny MM, Anmin M, Abozid H: Effects of antenatal magnesium sulfate therapy on cerebral and systemic hemodynamics in preterm newborns. Acta Obstet Gynecol Scand 2010;89:801-806.
7.
Doyle LW, Crowther CA, Middleton P, Marret S: Antenatal magnesium sulfate and neurologic outcome in preterm infants: a systematic review. Obstet Gynecol 2009;113:1327-1333.
8.
Conde-Agudelo A, Romero R: Antenatal magnesium sulfate for the prevention of cerebral palsy in preterm infants less than 34 weeks' gestation: a systematic review and metaanalysis. Am J Obstet Gynecol 2009;200:595-609.
9.
Cahill AG, Caughey AB: Magnesium for neuroprophylaxis: fact or fiction? Am J Obstet Gynecol 2009;200:590-594.
10.
Doyle LW: Antenatal magnesium sulfate and neuroprotection. Curr Opin Pediatr 2012;24:154-159.
11.
Nguyen TM, Crowther CA, Wilkinson D, Bain E: Magnesium sulphate for women at term for neuroprotection of the fetus. Cochrane Database Syst Rev 2013;2:CD009395.
12.
Levene M, Blennow M, Whitelaw A, Hanko E, Fellman V, Hartley R: Acute effects of two different doses of magnesium sulphate in infants with birth asphyxia. Arch Dis Child Fetal Neonatal Ed 1995;73:F174-F177.
13.
Ichiba H, Tamai H, Negishi H, Ueda T, Kim TJ, Sumida Y, Takahashi Y, Fujinaga H, Minami H: Randomized controlled trial of magnesium sulfate infusion for severe birth asphyxia. Pediatr Int 2002;44:505-509.
14.
Ichiba H, Yokoi T, Tamai H, Ueda T, Kim TJ, Yamano T: Neurodevelopmental outcome of infants with birth asphyxia treated with magnesium sulfate. Pediatr Int 2006;48:70-75.
15.
Groenendaal F, Rademaker CM, Toet MC, de Vries LS: Effects of magnesium sulphate on amplitude-integrated continuous EEG in asphyxiated term neonates. Acta Paediatr 2002;91:1073-1077.
16.
Rice JE 3rd, Vannucci RC, Brierley JB: The influence of immaturity on hypoxic-ischemic brain damage in the rat. Ann Neurol 1981;9:131-141.
17.
Dobbing J, Sands J: Comparative aspects of the brain growth spurt. Early Hum Dev 1979;3:79-83.
18.
Gunn AJ, Bennet L: Fetal hypoxia insults and patterns of brain injury: insights from animal models. Clin Perinatol 2009;36:579-593.
19.
Lorek A, Takei Y, Cady EB, Wyatt JS, Penrice J, Edwards AD, Peebles D, Wylezinska M, Owen-Reece H, Kirkbride V: Delayed (‘secondary') cerebral energy failure after acute hypoxia-ischemia in the newborn piglet: continuous 48-hour studies by phosphorus magnetic resonance spectroscopy. Pediatr Res 1994;36:699-706.
20.
Sameshima H, Ota A, Ikenoue T: Pretreatment with magnesium sulfate protects against hypoxic-ischemic brain injury but postasphyxial treatment worsens brain damage in seven-day-old rats. Am J Obstet Gynecol 1999;180:725-730.
21.
Turkyilmaz C, Turkyilmaz Z, Atalay Y, Soylemezoglu F, Celasun B: Magnesium pre-treatment reduces neuronal apoptosis in newborn rats in hypoxia-ischemia. Brain Res 2002;955:133-137.
22.
Cetinkaya M, Alkan T, Ozyener F, Kafa IM, Kurt MA, Koksal N: Possible neuroprotective effects of magnesium sulfate and melatonin as both pre- and post-treatment in a neonatal hypoxic-ischemic rat model. Neonatology 2011;99:302-310.
23.
Sameshima H, Ikenoue T: Long-term magnesium sulfate treatment as protection against hypoxic-ischemic brain injury in seven-day-old rats. Am J Obstet Gynecol 2001;184:185-190.
24.
Spandou E, Soubasi V, Papoutsopoulou S, Augoustides-Savvopoulou P, Loizidis T, Pazaiti A, Karkavelas G, Guiba-Tziampiri O: Neuroprotective effect of long-term MgSO4 administration after cerebral hypoxia-ischemia in newborn rats is related to the severity of brain damage. Reprod Sci 2007;14:667-677.
25.
Pazaiti A, Soubasi V, Spandou E, Karkavelas G, Georgiou T, Karalis P, Guiba-Tziampiri O: Evaluation of long-lasting sensorimotor consequences following neonatal hypoxic-ischemic brain injury in rats: the neuroprotective role of MgSO4. Neonatology 2009;95:33-40.
26.
Goni-de-Cerio F, Alvarez A, Lara-Celador I, Alvarez FJ, Alonso-Alconada D, Hilario E: Magnesium sulfate treatment decreases the initial brain damage alterations produced after perinatal asphyxia in fetal lambs. J Neurosci Res 2012;90:1932-1940.
27.
Thordstein M, Bagenholm R, Thiringer K, Kjellmer I: Scavengers of free oxygen radicals in combination with magnesium ameliorate perinatal hypoxic-ischemic brain damage in the rat. Pediatr Res 1993;34:23-26.
28.
Gunn AJ, Sirimanne ES, Gluckman PD: Magnesium sulfate therapy is not neuroprotective after moderate hypoxia-ischemia in the immature rat. Prenat Neonat Med 1996;1:155-158.
29.
Galvin KA, Oorschot DE: Postinjury magnesium sulfate treatment is not markedly neuroprotective for striatal medium spiny neurons after perinatal hypoxia/ischemia in the rat. Pediatr Res 1998;44:740-745.
30.
de Haan HH, Gunn AJ, Williams CE, Heymann MA, Gluckman PD: Magnesium sulfate therapy during asphyxia in near-term fetal lambs does not compromise the fetus but does not reduce cerebral injury. Am J Obstet Gynecol 1997;176:18-27.
31.
Penrice J, Amess PN, Punwani S, Wylezinska M, Tyszczuk L, D'Souza P, Edwards AD, Cady EB, Wyatt JS, Reynolds EO: Magnesium sulfate after transient hypoxia-ischemia fails to prevent delayed cerebral energy failure in the newborn piglet. Pediatr Res 1997;41:443-447.
32.
Greenwood K, Cox P, Mehmet H, Penrice J, Amess PN, Cady EB, Wyatt JS, Edwards AD: Magnesium sulfate treatment after transient hypoxia-ischemia in the newborn piglet does not protect against cerebral damage. Pediatr Res 2000;48:346-350.
33.
Gunn AJ, Gunn TR, de Haan HH, Williams CE, Gluckman PD: Dramatic neuronal rescue with prolonged selective head cooling after ischemia in fetal lambs. J Clin Invest 1997;99:248-256.
34.
Drury PP, Davidson JO, van den Heuij LG, Tan S, Silverman RB, Ji H, Blood AB, Fraser M, Bennet L, Gunn AJ: Partial neuroprotection by nNOS inhibition during profound asphyxia in preterm fetal sheep. Exp Neurol 2013;250C:282-292.
35.
Marinov MB, Harbaugh KS, Hoopes PJ, Pikus HJ, Harbaugh RE: Neuroprotective effects of preischemia intraarterial magnesium sulfate in reversible focal cerebral ischemia. J Neurosurg 1996;85:117-124.
36.
Sirin BH, Coskun E, Yilik L, Ortac R, Sirin H, Tetik C: Neuroprotective effects of preischemia subcutaneous magnesium sulfate in transient cerebral ischemia. Eur J Cardiothorac Surg 1998;14:82-88.
37.
Lee EJ, Ayoub IA, Harris FB, Hassan M, Ogilvy CS, Maynard KI: Mexiletine and magnesium independently, but not combined, protect against permanent focal cerebral ischemia in Wistar rats. J Neurosci Res 1999;58:442-448.
38.
Westermaier T, Hungerhuber E, Zausinger S, Baethmann A, Schmid-Elsaesser R: Neuroprotective efficacy of intra-arterial and intravenous magnesium sulfate in a rat model of transient focal cerebral ischemia. Acta Neurochir (Wien) 2003;145:393-399.
39.
Zhou H, Ma Y, Zhou Y, Liu Z, Wang K, Chen G: Effects of magnesium sulfate on neuron apoptosis and expression of caspase-3, bax and bcl-2 after cerebral ischemia-reperfusion injury. Chin Med J (Engl) 2003;116:1532-1534.
40.
Chung SY, Lin JY, Lin MC, Liu HM, Wang MF, Cheng FC: Synergistic efficacy of magnesium sulfate and FK506 on cerebral ischemia-induced infarct volume in gerbil. Med Sci Monit 2004;10:BR105-BR108.
41.
Izumi Y, Roussel S, Pinard E, Seylaz J: Reduction of infarct volume by magnesium after middle cerebral artery occlusion in rats. J Cereb Blood Flow Metab 1991;11:1025-1030.
42.
Yang Y, Li Q, Ahmad F, Shuaib A: Survival and histological evaluation of therapeutic window of post-ischemia treatment with magnesium sulfate in embolic stroke model of rat. Neurosci Lett 2000;285:119-122.
43.
Blair JL, Warner DS, Todd MM: Effects of elevated plasma magnesium versus calcium on cerebral ischemic injury in rats. Stroke 1989;20:507-512.
44.
Zhu H, Meloni BP, Moore SR, Majda BT, Knuckey NW: Intravenous administration of magnesium is only neuroprotective following transient global ischemia when present with post-ischemic mild hypothermia. Brain Res 2004;1014:53-60.
45.
Zhu H, Meloni BP, Bojarski C, Knuckey MW, Knuckey NW: Post-ischemic modest hypothermia (35 degrees C) combined with intravenous magnesium is more effective at reducing CA1 neuronal death than either treatment used alone following global cerebral ischemia in rats. Exp Neurol 2005;193:361-368.
46.
Campbell K, Meloni BP, Knuckey NW: Combined magnesium and mild hypothermia (35 degrees C) treatment reduces infarct volumes after permanent middle cerebral artery occlusion in the rat at 2 and 4, but not 6 h. Brain Res 2008;1230:258-264.
47.
Song W, Wu YM, Ji Z, Ji YB, Wang SN, Pan SY: Intra-carotid cold magnesium sulfate infusion induces selective cerebral hypothermia and neuroprotection in rats with transient middle cerebral artery occlusion. Neurol Sci 2013;34:479-486.
48.
Milani H, Lepri ER, Giordani F, Favero-Filho LA: Magnesium chloride alone or in combination with diazepam fails to prevent hippocampal damage following transient forebrain ischemia. Braz J Med Biol Res 1999;32:1285-1293.
49.
Schanne FA, Gupta RK, Stanton PK: 31P-NMR study of transient ischemia in rat hippocampal slices in vitro. Biochim Biophys Acta 1993;1158:257-263.
50.
Garnier Y, Middelanis J, Jensen A, Berger R: Neuroprotective effects of magnesium on metabolic disturbances in fetal hippocampal slices after oxygen-glucose deprivation: mediation by nitric oxide system. J Soc Gynecol Investig 2002;9:86-92.
51.
Tsuda T, Kogure K, Nishioka K, Watanabe T: Mg2+ administered up to twenty-four hours following reperfusion prevents ischemic damage of the CA1 neurons in the rat hippocampus. Neuroscience 1991;44:335-341.
52.
Khashaba MT, Shouman BO, Shaltout AA, Al-Marsafawy HM, Abdel-Aziz MM, Patel K, Aly H: Excitatory amino acids and magnesium sulfate in neonatal asphyxia. Brain Dev 2006;28: 375-379.
53.
Oppelt WW, MacIntyre I, Rall DP: Magnesium exchange between blood and cerebrospinal fluid. Am J Physiol 1963;205:959-962.
54.
Hallak M, Berman RF, Irtenkauf SM, Evans MI, Cotton DB: Peripheral magnesium sulfate enters the brain and increases the threshold for hippocampal seizures in rats. Am J Obstet Gynecol 1992;167:1605-1610.
55.
Westermaier T, Stetter C, Kunze E, Willner N, Raslan F, Vince GH, Ernestus RI: Magnesium treatment for neuroprotection in ischemic diseases of the brain. Exp Transl Stroke Med 2013;5:6.
56.
Altura BM, Altura BT, Carella A, Gebrewold A, Murakawa T, Nishio A: Mg2+-Ca2+ interaction in contractility of vascular smooth muscle: Mg2+ versus organic calcium channel blockers on myogenic tone and agonist-induced responsiveness of blood vessels. Can J Physiol Pharmacol 1987;65:729-745.
57.
Perales AJ, Torregrosa G, Salom JB, Miranda FJ, Alabadi JA, Monleon J, Alborch E: In vivo and in vitro effects of magnesium sulfate in the cerebrovascular bed of the goat. Am J Obstet Gynecol 1991;165:1534-1538.
58.
Heidarianpour A, Sadeghian E, Gorzi A, Nazem F: The influence of oral magnesium sulfate on skin microvasculature blood flow in diabetic rats. Biol Trace Elem Res 2011;143:344-350.
59.
Haddy FJ: Local effects of sodium, calcium and magnesium upon small and large blood vessels of the dog forelimb. Circ Res 1960;8:57-70.
60.
DeBow SB, Clark DL, MacLellan CL, Colbourne F: Incomplete assessment of experimental cytoprotectants in rodent ischemia studies. Can J Neurol Sci 2003;30:368-374.
61.
Geddes R, Vannucci RC, Vannucci SJ: Delayed cerebral atrophy following moderate hypoxia-ischemia in the immature rat. Dev Neurosci 2001;23:180-185.
62.
Schmid-Elsaesser R, Hungerhuber E, Zausinger S, Baethmann A, Reulen HJ: Combination drug therapy and mild hypothermia: a promising treatment strategy for reversible, focal cerebral ischemia. Stroke 1999;30:1891-1899.
Copyright / Drug Dosage / Disclaimer
Copyright: All rights reserved. No part of this publication may be translated into other languages, reproduced or utilized in any form or by any means, electronic or mechanical, including photocopying, recording, microcopying, or by any information storage and retrieval system, without permission in writing from the publisher.
Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug.
Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.