Skip to main content
Erschienen in: Acta Neuropathologica 4/2018

Open Access 16.03.2018 | Review

Energy metabolism in ALS: an underappreciated opportunity?

verfasst von: Tijs Vandoorne, Katrien De Bock, Ludo Van Den Bosch

Erschienen in: Acta Neuropathologica | Ausgabe 4/2018

Abstract

Amyotrophic lateral sclerosis (ALS) is a relentlessly progressive and fatal neurodegenerative disorder that primarily affects motor neurons. Despite our increased understanding of the genetic factors contributing to ALS, no effective treatment is available. A growing body of evidence shows disturbances in energy metabolism in ALS. Moreover, the remarkable vulnerability of motor neurons to ATP depletion has become increasingly clear. Here, we review metabolic alterations present in ALS patients and models, discuss the selective vulnerability of motor neurons to energetic stress, and provide an overview of tested and emerging metabolic approaches to treat ALS. We believe that a further understanding of the metabolic biology of ALS can lead to the identification of novel therapeutic targets.

Introduction

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder characterized by the selective and progressive degeneration of motor neurons in the brain and spinal cord. Motor neuron deterioration leads to muscle weakness and results in death due to respiratory failure typically within 3–5 years after diagnosis [25]. In the Western world, the lifetime risk of developing ALS is estimated to be 1 in 400 [89].
ALS is a highly heterogeneous disease [187]; 5–10% of patients have a familial form in which inheritance almost exclusively occurs via an autosomal dominant Mendelian pattern. While over 120 potential ALS genes (http://​alsod.​iop.​kcl.​ac.​uk/​) have been identified, more than half of familial ALS patients carry mutations in either ‘superoxide dismutase 1’ (SOD1), ‘TAR DNA binding protein’ (TARDBP), ‘fused in sarcoma’ (FUS), or carry a hexanucleotide repeat expansion in an intronic region of the ‘chromosome 9 open reading frame 72’ (C9ORF72) gene [190]. Despite the genetic heterogeneity, most patients show cytoplasmic inclusions in motor neurons which stain positive for TDP-43, the protein product of TARDBP [136]. This suggests that similar pathogenic mechanisms may be present in different ALS subtypes. Although most ALS patients have no family history, unraveling the genetic basis of the disease led to an array of ALS models, put forth different processes believed to be involved in ALS pathogenesis, and led to various clinical trials [190]. Despite these efforts, translation of preclinical findings into effective therapeutic strategies remained poor. Riluzole and edaravone are the only FDA-approved drugs to treat ALS. Riluzole prolongs life by only a few months [12] and edaravone improves patient functionality scores in a subset of patients [165, 211]. Due to the unavailability of effective drugs, there is an urgent need for new treatment modalities in ALS.
A growing body of evidence shows dysregulated energy metabolism in ALS patients and models. Several of the metabolic abnormalities in ALS correlate to disease susceptibility and progression. Moreover, the remarkable vulnerability of motor neurons to energy depletion has become increasingly clear. In this review, we focus on how energy metabolism is impaired in ALS, and how motor neuron physiology contributes to their particular vulnerability to metabolic stress. We also discuss tested and emerging metabolism-centric therapeutic avenues for ALS.

Systemic metabolism correlates to ALS disease course

Control of whole-body energy homeostasis, the balance between energy uptake and expenditure, is crucial to maintain stable body weight and hence overall health [105]. In ALS patients, energy homeostasis is imbalanced [57]. While energy uptake is often lowered [1], energy expenditure is suggested to be increased in a significant proportion of patients with ALS [21]. While this observation stems from predictive equations which still need validation in ALS patients and should, therefore, be interpreted with care [176], energy expenditure exceeds uptake in most ALS patients, leading to reduced fat depots [81]. Imbalanced energy homeostasis is also a consistent finding in different SOD1 [56, 62] and TDP-43 mouse models [30, 36]. Recently, the melanocortin pathway, a critical regulator of energy homeostasis and food intake in the hypothalamus [184], was hypothesized to contribute to imbalanced energy homeostasis in ALS patients [67] and mice [201]. However, reducing energy expenditure and inducing hyperphagia by targeting this pathway in mutant SOD1G93A mice did not improve motor function or lifespan [53]. While the cause and importance of dysregulated energy homeostasis in human ALS remains to be established, body weight loss is an important prognostic factor in patients [149]. A lower pre-symptomatic body mass index has been reported in ALS patients [86, 126, 149] and the ALS risk is reduced up to 40% among obese individuals [138]. In agreement, increased prediagnostic body fat [65], subcutaneous fat [111], and serum leptin [135] were associated with a decreased risk of ALS mortality.
The majority of ALS patients suffer from hypolipidemia [215]. Of note, hypolipidemia is also present in mutant SOD1 mice [62, 98] and precedes clinical onset in mutant SOD1G93A mice [98]. Whether hypolipidemia is also a preclinical feature in human ALS patients is difficult to assess, since diagnostic certainty is only reached in a progressed stage of the disease. In addition, elevated serum cholesterol and apolipoprotein E levels prolong survival and delay disease progression in ALS patients in most [52, 54, 103], but not all [31], studies, while statin treatment was associated with worsened outcome [224]. An additional study showed a positive correlation between blood lipids and respiratory function in ALS patients, potentially due to the decrease in CO2 production, which lowers the load on ventilatory muscles [27, 31].
Interestingly, ALS patients suffering from diabetes show a delay in the onset of motor symptoms for up to 4 years [87]. A large case–control study reported an estimated odds ratio for ALS association with diabetes of 0.61 (95% confidence interval: 0.46–0.80) [99]. Remarkably, type II diabetes was associated with a decreased risk of ALS (odds ratio 0.79, 95% confidence interval: 0.68–0.91) [127], while type I diabetes was associated with an increased risk (odds ratio 5.38, 95% confidence interval: 1.87–15.51) [194]. These data suggest that a potential protective effect is restricted to type II diabetes. Large longitudinal studies are required to determine whether insulin resistance (a hallmark of type II diabetes) per se has a protective effect against ALS or whether the protective effect is secondary to environmental and/or genetic factors that contribute to the development of type II diabetes. Moreover, ALS patients often develop insulin resistance during the course of the disease [154]. Since muscle tissue represents the major site of glucose consumption and storage, the development of insulin resistance during ALS is considered a consequence of muscle atrophy, although molecular evidence is still lacking. Even more, it has been suggested that deregulation of carbohydrate metabolism might contribute to ALS pathogenesis (see below).
Altogether, systemic metabolic defects in ALS correlate with disease duration and/or progression [21, 26, 81]. However, it remains to be determined whether and how these defects are causally connected to ALS pathogenesis.

Motor neuron metabolism in health

Since its first description by Charcot in 1869, the characteristic selective degeneration and death of motor neurons in ALS has remained an enigma. Neurons are large, polarized, excitable cells and, therefore, face unique challenges to maintain energy homeostasis (Fig. 1). They are the main contributors to the impressive energy demand of the central nervous system (CNS). First, action potential propagation is highly dependent on the Na+/K+-ATPase [75]. Second, due to the extensive length of their neurites, neurons, and, a fortiori, motor neurons, depend on axonal transport [155]. Importantly, the molecular motors driving axonal transport hydrolyze one ATP molecule, generated via on-board glycolysis [217], for every 8-nm displacement of their cargo [79, 167]. Since synapses are major sites of neuronal energy consumption, the trafficking of mitochondria is critical to meet synaptic energy requirements [174]. On top of this, high ATP concentrations are needed to keep proteins soluble [143]. The high dependence of motor neurons on continuous energy provision to maintain their normal function and integrity renders them particularly vulnerable to energetic stress [106].
To meet its substantial energy demand, the CNS largely relies on glucose as an energy substrate [121]. Recent in vitro and ex vivo studies have indicated though that neurons can readily oxidize several non-glucose substrates and that a switch towards glutamate oxidation could protect neurons from excitotoxic cell death [49, 61]. These data, nonetheless, require in vivo confirmation, since the absence of the blood–brain/spinal cord barrier and specific conditions of the CNS microenvironment might make it difficult to translate in vitro findings to an in vivo situation. Indeed, to date, the evidence indicates that only ketone bodies can sustain the energetic requirements of the CNS in conditions of severe glucose deprivation [35, 104]. Fatty acids are only poorly used as an energy substrate presumably due to: the slow passage of fatty acids across the blood–brain and blood–spinal cord barrier, the higher oxygen cost of fatty acid oxidation, the elevated superoxide generation during fatty acid oxidation in combination with poor anti-oxidant defense mechanisms of neurons, and the slower rate of ATP generation of fatty acid oxidation [168]. More than 90% of ATP generation in the CNS occurs via mitochondrial oxidative phosphorylation [82]. Acute fluctuations in ATP demand in the CNS are met by the creatine/phosphocreatine system, which represents an instant way to liberate high-energy phosphates for ATP by the transphosphorylation of phosphocreatine by creatine kinases [4]. Since ATP turnover in the CNS is high and substrate reserves small, the creatine/phosphocreatine system is crucial to buffer ATP fluctuations upon neuronal firing [16]. Moreover, the faster diffusion rate of phosphocreatine compared to ATP, and creatine compared to ADP [200], makes the creatine/phosphocreatine system suitable to connect sites of ATP generation to sites of ATP consumption.
Despite glucose being the dominant energy substrate, the CNS is a highly heterogeneous tissue composed of different cell types which show distinct metabolic profiles (Fig. 2). Differences are mainly studied in neurons and astrocytes, and indicate that neurons are predominantly oxidative and that astrocytes are predominantly glycolytic [22, 121, 219]. Under normal conditions, carbohydrate catabolism comprises the conversion of glucose to pyruvate via glycolysis followed by the full oxidation of glucose, or its metabolites pyruvate or lactate, in the mitochondria by the tricarboxylic acid (TCA) cycle and electron transport chain. Oxidative catabolism requires oxygen and generates 31–36 molecules of ATP for every molecule of glucose (or half of it if lactate or pyruvate is used as substrate). However, when the availability of oxygen is low (or in specific cell types—see below), glucose is only glycolytically catabolized to pyruvate, which generates only two molecules of ATP for each molecule of glucose, and is subsequently converted to lactate. This is a necessary step, since the regeneration of nicotinamide–adenine dinucleotide (NAD+) is required to keep glycolysis going when oxygen is limited [116]. Pyruvate dehydrogenase (PDH) is crucial to allow pyruvate entry into the TCA cycle and, hence, controls oxidative versus anaerobic catabolism. In astrocytes, PDH activity is low compared to neurons [73]; and pyruvate dehydrogenase kinase 4 expression, the main kinase suppressing PDH activity, is high, leading to higher glycolysis and lactate production [219]. In contrast, neurons have a lower rate of glycolysis under normal conditions due to the constant degradation of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3), a key positive modulator of glycolysis, by the E3 ubiquitin ligase anaphase-promoting complex/cyclosome [18, 78]. In agreement, glia transport and metabolize glucose analogues faster than neurons both ex vivo [9] and in vivo [85]. In addition, a part of the glucose that is taken up by neurons does not enter glycolysis but is instead directed to the pentose phosphate pathway (PPP) during which the anti-oxidant reduced glutathione is regenerated (Fig. 2). Both overexpression [78] and stabilization [158] of PFKFB3 in neurons activated glycolysis at the expense of the PPP and resulted in oxidative stress and apoptotic death. These data suggest that neuronal homeostasis is particularly dependent on a tight balance between glycolysis and PPP flux to ensure sufficient ATP production while maintaining anti-oxidant status.
The more oxidative profile of neurons and more glycolytic profile of glia becomes more pronounced upon neuronal activity [76, 124], suggesting that other energy substrates are used to meet the neuronal energy demand during neuronal activity. Lactate is consumed in an activity-dependent manner in the CNS [162, 212] and is mainly oxidized by neurons compared to astrocytes [20, 196]. According to the astrocyte-neuronal lactate shuttle hypothesis, lactate is provided to neurons by astrocytes [147]. In brief, the reuptake of glutamate by astrocytes depletes their ATP stores, which stimulates the uptake of glucose and subsequently the glycolytic flux. The resulting lactate is mainly exported through the astrocyte-specific monocarboxylate transporter 4 (MCT4) and taken up by the neuron-specific MCT2 transporters. Next, it is fully oxidized to generate ATP (for a review, see [121]). In addition to lactate, astrocytes can also provide pyruvate and ketone bodies to the neurons [148]. Oxidative phosphorylation of glia-derived substrates in neurons leads to the generation of reactive oxygen species (ROS) which promote lipid production [112, 113]. Those lipids are transported to astrocytes via a fatty acid transport proteins (FATP) and apolipoprotein-dependent mechanism where they can form lipid droplets, be shunted into the ketogenic pathway, or undergo fatty acid oxidation [71] (Fig. 2). Impaired transport of lipids from neurons to glia accelerates neurodegeneration [112], suggesting a pro-survival function of neuron-derived lipids in glia by serving as in situ energy substrates under stress. Of note, while astrocyte-neuron metabolic coupling seems to be essential for nervous system homeostasis in Drosophila [204] and mice [64, 109, 120], it does not imply a complete metabolic compartmentalization of glycolysis versus oxidative phosphorylation in glia and neurons, respectively [7, 48]. Indeed, neurons also take up and metabolize glucose and increase glucose consumption in an activity-dependent manner [6, 47, 144]. In addition, neurons can catabolize glucose and lactate at the same time [115]. Therefore, it is likely that both oxidative phosphorylation of glia-derived energy substrates as well as neuronal glycolysis contribute to ATP production in high-energy demanding cellular situations.
Astrocyte contact with neurons is generally limited to the neuronal soma, synapses, and nodes of Ranvier, leaving the largest part of the axon without metabolic support from astrocytes. This is especially true for motor neurons. In contrast, oligodendrocytes are well connected to the axon and perfectly positioned to support the metabolic demands of neurons [151]. These glial cells highly express MCT1, which is the MCT with the highest affinity for lactate [152]. MCT1 inhibition in organotypic spinal cord slice cultures reduced motor neuron survival, but this effect was rescued by the addition of high concentrations of lactate to the culture medium [109], suggesting that oligodendrocyte-derived lactate contributes to the survival of motor neurons. In addition, astrocyte-to-oligodendrocyte coupling is essential for myelination [193]. Whether coupling between different glial cells is also involved in the metabolic support of motor neurons is unknown.
In summary, motor neurons require vast amounts of energy while having limited energy stores. Therefore, neuronal function and survival requires the continuous provision of substantial amounts of nutrients for ATP production. Under normal conditions, neurons are predominantly oxidative and astrocytes are predominantly glycolytic [219]. In addition, neurons keep a tight balance between glycolysis and flux through the PPP to maintain their anti-oxidant status while ensuring optimal ATP production. Of note, the metabolic characteristics of neurons have been studied to a large extent in cortical neurons. Whether and how motor neurons, due to their specific anatomy and microenvironment, have different metabolic properties, remain to be determined. Their high need for continuous energy provision, nonetheless, renders motor neurons particularly vulnerable to energetic stress [106]; and this could contribute to the selective vulnerability and degeneration of motor neurons observed in ALS. Indeed, fast-fatigable motor neurons, which have the highest peak needs of ATP [106], are initially targeted and are more severely affected during ALS compared to slow motor neurons [137].

Motor neuron metabolism in ALS

Cellular energy homeostasis is impaired in ALS

Mammalian AMP-activated protein kinase (AMPK) is a major cellular energy sensor activated by falling energy status. Upon activation, AMPK restores energy homeostasis by promoting catabolic pathways, resulting in ATP generation, and inhibiting anabolic pathways that consume ATP [74]. Enhanced AMPK activation was observed in motor neurons of ALS patients and correlated closely with the extent of cytoplasmic mislocalization of TDP-43 [114]. In NSC34 motor neuron-like cells, 5-aminoimidazole-4-carboxamide-1-β-d-ribofuranoside (AICAR)-mediated activation of AMPK caused TDP-43 mislocalization [114]. These data link energy depletion in human motor neurons to ALS-related TDP-43 pathology. AMPK activation was also increased in spinal cord cultures or lysates of mutant SOD1G93A mice [110]. Pharmacological activation of AMPK worsened disease outcome in these mice [91]. In accordance, reducing AMPK-activity improved disease outcome in vitro or in C. elegans models expressing mutant SOD1 or TDP-43 [110]. These studies collectively show disturbed energy homeostasis at the cellular level in ALS and demonstrate its role in TDP-43 proteinopathy, the histopathological signature of ALS. While a clear mechanistic link between AMPK activation and TDP-43 mislocalization is currently lacking, nucleocytoplasmic transport is known to be an energy-dependent process [17]. It is, therefore, possible that cytoplasmic mislocalization of an aggregation prone protein such as TDP-43 [88] results from AMPK-mediated inhibition of nucleocytoplasmic transport.

Mitochondrial dysfunction, an ALS hallmark

Mitochondrial dysfunction is a clinical hallmark of both sporadic and familial ALS [23, 55, 163]. As a consequence, multiple processes in which mitochondria play a key role are extensively investigated in ALS [182]. Seminal studies have shown dense clusters of mitochondria in the anterior horn of the lumbar spinal cord [164] and presynaptic mitochondrial swelling in motor neurons [180] of ALS patients. In addition, the amount of mitochondrial DNA, a direct marker of mitochondrial abundance, was reduced in the spinal cord from familial and sporadic ALS patients [207]. In mice carrying the SOD1G37R mutation, membrane-bound vacuoles derived from degenerating mitochondria were observed in neurites [210]. Massive mitochondrial degeneration in motor neurons of mutant SOD1G93A mice was already observed at disease onset [41, 102]. The observation that mitochondrial morphology is also abnormal in various murine FUS [80, 183] and TDP-43 models [170, 213] is important, since overexpression of human SOD1 per se, rather than the pathogenic effect of the mutation, induces mitochondrial vacuolization [84]. Besides morphological abnormalities, functional changes are present in ALS mitochondria. In spinal cord mitochondria from ALS patients, there was decreased activity of the electron transport chain (ETC) complexes I + III, II + III, and IV [207]. Decreased activity of the mitochondrial enzymes citrate synthase and cytochrome c oxidase was also reported in motor neurons from ALS patients [19]. Furthermore, impaired activities of complex I + III, II + III, and IV were also observed in mutant SOD1G93A mice [129]. Importantly, reduced respiration and ATP synthesis preceded behavioral deficits in mutant SOD1G93A mice [90, 129, 188], indicating a role in pathology. This has not yet been validated in other ALS models.
In addition to abnormal mitochondrial morphology and function, the cellular distribution of mitochondria is altered in ALS. In ALS patients, mitochondrial accumulation was observed in the cell body and proximal axon hillock [163]. Disturbed mitochondrial dynamics were also observed in embryonic and adult motor neurons expressing mutant SOD1G93A [15, 45, 185], mutant TDP-43 overexpressing mice [122], and FUS patient-derived motor neurons [70]. Expressing mutant TDP-43 in motor neurons also induced aberrant mitochondrial distribution [205]. Miro1, a mitochondrial outer membrane protein coupling mitochondria to the axonal transport machinery, is downregulated in ALS, suggesting a mechanistic basis for impaired mitochondrial distribution in ALS [134, 218].
Abnormal mitochondrial physiology is a consistent observation in ALS patients and in multiple ALS models (Fig. 3). In mutant SOD1 [90, 129, 188] and FUS [183] ALS mouse models, mitochondrial dysfunction is an early event and overexpressing peroxisome proliferator activated receptor-gamma coactivator 1 alpha (PGC1α), a major regulator of mitochondrial biogenesis, improved survival, motor function, and motor neuron survival in mutant SOD1G93A mice [221]. PGC1α expression is also downregulated in the CNS of FUS-ALS mice and FUS patient derived motor neurons [10]. Therefore, improving mitochondrial biogenesis may be an attractive therapeutic strategy for ALS. It should be noted that mitochondrial abnormalities in ALS are not restricted to motor neurons. In skeletal muscle from patients, mitochondria are also structurally [130, 203] and functionally [38, 58] abnormal. While alterations in skeletal muscle can affect neuromuscular junction integrity [209], their role in ALS remains controversial (for a review, see [117]).
This said, the underlying cause of mitochondrial dysfunction in ALS and whether mitochondrial dysfunction is causally linked to motor neuron pathology in ALS still remains an open question. It is also not yet clear whether mitochondrial defects are present in all ALS subtypes. Moreover, mechanisms linking mutations in TDP-43 and FUS to aberrant mitochondrial physiology remain to be determined. Overall, the current literature suggests that aberrant mitochondrial physiology could be an important contributing factor to the pathogenesis of ALS.

Carbohydrate metabolism and ALS

In line with mitochondrial dysfunction, glucose uptake in the motor-sensory cortex of ALS patients is reduced [77, 140]. FDG-PET studies linked the reduction of glucose uptake and phosphorylation to the severity of the disease [42]. Another FDG-PET study consisting of 81 patients with a suspected diagnosis of ALS was able to correctly classify 95% of ALS cases, indicating reduced glucose uptake as an early diagnostic event in ALS [197]. In mutant SOD1G93A mice, glucose uptake in the spinal cord increased pre-symptomatically, but declined progressively during disease progression [133]. Under physiological conditions, there is a tight coupling between blood flow and glucose metabolism in the CNS [66]. Remarkably, the pre-symptomatic increase in glucose uptake in the spinal cord of mutant SOD1G93A mice was not matched by increases in spinal blood flow [133]. Despite reduced glucose uptake, the spinal cord from end-stage mutant SOD1G93A mice, as well as from autopsied ALS patients, is characterized by elevated concentrations of glycogen [50]. Blood flow–metabolism uncoupling together with increased glycogen storage in the CNS suggests a decreased ability to catabolize carbohydrates in mutant SOD1G93A mice. However, it is debated whether reduced glucose uptake in the CNS of ALS patients reflects a reduction in neuronal carbohydrate catabolism or a reduction in the number of motor neurons. Nevertheless, the ability to catabolize carbohydrates appears to be reduced in human ALS patients as the expression of phosphoglucomutase 2 like 1 and phosphoglycerate kinase, two key enzymes in glycolysis, is downregulated in fibroblasts from sporadic ALS patients [157]. In agreement, a recent proteomic study in sporadic ALS skin fibroblasts showed a marked reduction in components of glycolysis [188]. Whole-genome expression profiling in the motor cortex of sporadic ALS patients also showed a significant downregulation of glycolytic genes [108]. Another study in post-mortem cortex of ALS patients identified an over twofold reduction in PFKFB3 mRNA content [206]. In contrast, introducing mutant SOD1 in human fibroblasts or NSC34 motor neuron-like cells increased glycolysis and reduced mitochondrial ATP generation [3, 195]. Given the limited capacity of neurons to upregulate glycolysis [78, 158], the physiological relevance to ALS of the upregulation of glycolysis in these cells remains to be established. Nevertheless, neurons can upregulate glycolysis [47] and oxidative stress is evident in post-mortem samples of ALS patients [63]. It is, therefore, possible that during severe energetic stress, motor neurons sacrifice their redox status to alleviate energetic stress and eventually die due to excessive oxidative stress.
It remains to be determined whether these metabolic alterations are taking place in motor neurons or glia (Fig. 3). Given that in both the cortex and the spinal cord, motor neurons represent a quantitatively minor population, functional metabolic studies on induced pluripotent stem cell (iPSC)-derived motor neurons from patients could provide valuable insight. Compared to neurons, glia are more glycolytic, and, therefore, likely significantly contribute to the observed reductions in transcription level of key glycolytic transcripts in the CNS of ALS patients. In oligodendrocytes from ALS patient and mutant SOD1G93A mice, the expression of MCT1 transporters is downregulated [92, 109, 150]. In addition, the downregulation of the glutamate transporter GLT-1 in astrocytes from ALS patients is well established [161]. These observations suggest that the reduced glycolytic capacity is, at least in part, due to changes in glial metabolism.
Taken together, reductions in glucose uptake and increased glycogen storage in the CNS of ALS patients and ALS animal models suggest a reduced capacity to catabolize glucose. Noteworthy, riluzole enhanced CNS glucose uptake both in vitro [43] and in vivo [32], suggesting that improving glucose transport rates in ALS affected cells may be a potential therapeutic avenue. While studies evaluating specific pathways are scarce, glycolysis seems to be downregulated. How different cell types in the CNS contribute to reduced carbohydrate catabolism remains to be investigated. Therefore, future studies investigating the metabolic fate of glucose, using, i.e., traceable glucose analogues [8], in ALS models are urgently needed. Moreover, it is not clear how reduced carbohydrate catabolism might affect neuronal function.

Metabolic treatments tested in ALS

Energy metabolism is altered in ALS (Fig. 3) and correlates to disease progression, suggesting a role for energy metabolism in ALS pathogenesis. As a consequence, targeting metabolism represents a rational strategy to treat ALS. Below, we will give an overview of therapeutic approaches that target energy metabolism and have been tested in ALS patients and/or preclinical models (for an overview, see Table 1). Most approaches focus on increasing the provision of energetic substrates or improving mitochondrial function. Some strategies target the electron transport chain as the most important cellular source of oxidative stress. Creatine has also been investigated for its energy buffering capacities. Of note, most metabolic treatments have multiple mechanisms of action. One example is dichloroacetate, which improves mitochondrial function indirectly by stimulating the conversion of pyruvate to acetyl coenzyme A (ACoA), and, therefore, also provides additional energy substrates to the TCA cycle. For reasons of clarity, we classified treatments according to their principal mechanism of action.
Table 1
Metabolic treatments tested in ALS
Putative mechanism of action
Metabolic treatment
Effect on ALS models
Effect on ALS patients
Energy buffering and transport
Creatine
Improved lifespan, motor neuron survival, and motor function in mutant SOD1G93A mice [87]
No efficacy in phase II/III clinical trials [55, 139, 151]
Oxidative stress
Coenzyme Q10
Improved survival in mutant SOD1G93A mice—[111]
No efficacy in phase II clinical trial [81]
MitoQ
Reduced toxicity of mutant SOD1G93A rat astrocytes to healthy motor neurons in co-culture [24]
Improved motor function, survival, and histopathology in mutant SOD1G93A mice [114]
To be tested
Dexpramipexole
Improved survival, and motor function in mutant SOD1G93A mice in one study [36], but not in a second study [177]
No effect in patient derived iPSCs [189]
No effect in rat cortical neurons transfected with mutant or wild-type TDP-43 [177]
No efficacy in phase III clinical trial [33]
Edaravone
Delayed motor neuron degeneration and spinal cord SOD1 deposition in mutant SOD1G93A mice [71]
Delayed disease progression in wobbler mice [69]
Improved motor performance in mutant SOD1H46R rats [5]
Efficacy in a subset of ALS patients [184], FDA-approved
Additional and/or alternative fuel
High caloric diet
Delayed disease onset and extended survival in mutant SOD1G93A, mutant SOD1G86R, and mutant TDP-43A315T mice [30, 46]
Delayed motor neuron loss in the spinal cord of mutant SOD1G93A mice [46]
Promising results in a phase II clinical trial [182]
Ketone bodies
Ketogenic diets delay disease onset, improved motor neuron survival but not lifespan in mutant SOD1G93A mice [195]
Ketone esters are to be tested in ALS models
To be tested
Medium-chain triglycerides
Delayed disease onset, and improved motor neuron survival in mutant SOD1G93A mice [167, 193]
To be tested
Pyruvate
Improved motor performance, disease progression, and lifespan in mutant SOD1G93A mice [121] but not in a subsequent study [48]
To be tested
Mitochondrial function
Dichloroacetate
Improved survival, delayed disease onset, and improved motor neuron survival in mutant SOD1G93A mice [113, 120]
To be tested
Acetyl-l-carnitine
Neurotrophic effects in rat embryonic motor neurons [10]
Improved survival in mutant SOD1G93A mice [86]
Promising results in a phase II clinical trial [8]

Creatine

As described above, the creatine/phosphocreatine system plays a crucial role in neurons for cellular energy buffering and transport [4]. In mutant SOD1G93A mice, creatine treatment prevented ATP depletion in the cerebellar cortex and spinal cord [26], but not in skeletal muscle [46]. Creatine treatment in mutant SOD1G93A mice markedly improved motor neuron survival but only moderately enhanced motor function and lifespan [101]. No studies evaluated the effect of creatine in other ALS mouse models. In ALS patients, creatine supplementation did not affect survival or disease progression [69, 160, 173]. Several reasons can explain the different effect of creatine in mutant SOD1G93A mice and ALS patients. First, the efficacy of creatine in mutant SOD1G93A mice can, at least in part, be explained by a buffering effect of creatine on SOD1 overexpression-related mitochondriopathy [13, 84]. In addition, the treatment was initiated at different disease stages in mice compared to patients. While creatine intake in mutant SOD1G93A mice was started before symptom onset and before the reduction in ATP concentration in the CNS [26], ALS patients’ treatment started at least 1 year after symptom onset when already extensive alterations in energy metabolism are present. While creatine supplementation is able to prevent neuronal ATP depletion in some conditions and for short periods of time [186], it acts by energy buffering and transport without contributing to ATP production [4]. Therefore, it is likely that when treatment only commences during progressed disease states, the cascade of metabolic dysfunction is too far advanced for interventions to be successful.

Targeting oxidative stress

The interest in the role of oxidative stress was nurtured for decades by the finding that mutations in SOD1 can cause ALS [159]. Whether oxidative stress is a primary or secondary disease mechanism in human ALS is still unclear. The recent discovery that the free radical scavenger edaravone improves ALS functional rating scale (ALSFRS) scores of a subgroup of ALS patients suggests that oxidative stress affects motor neuron death [211]. Nevertheless, most clinical trials targeting oxidative stress failed to demonstrate clinical efficacy (see below). Given the vulnerability of motor neurons to oxidative stress [172], neurons employ different strategies to minimize ROS accumulation [168]. Hence, oxidative stress might be an indicator of advanced cellular damage rather than an early pathological event. Identifying the exact underlying mechanism responsible for the efficacy of edaravone in ALS patient subpopulations could provide further insight in the role of oxidative stress in ALS.

Coenzyme Q10 and MitoQ

Coenzyme Q, also known as ubiquinone, is the only endogenous lipid-soluble anti-oxidant found in humans. It acts as an essential cofactor in the electron transport chain where it accepts electrons from complex I and II and shuttles them to complex III. Its quinone group can be reduced to quinol, explaining its anti-oxidative properties [37]. In mutant SOD1G93A mice, coenzyme Q10 induced a mild improvement in survival [128]. However, a phase II clinical trial, treating ALS patients with small amounts of coenzyme Q10 for 9 months, did not observe improvements on ALSFRS [95]. Of note, the feeding regimen in this study only induced a moderate increase in plasma coenzyme Q10 levels and the previous studies in Parkinson’s disease suggest that doses up to 100-fold of the dose used are needed to slow disease progression [178, 179]. Although administration of high doses of exogenous coenzyme Q10 is well tolerated in humans [178], its hydrophobicity compromises bioavailability [128]. To improve this, MitoQ, a mitochondrion-targeted and recyclable coenzyme Q10 analogue, was developed [96]. MitoQ showed enhanced bioavailability and improved mitochondrial function in different neuronal cell types exposed to oxidative stress [189]. Rat mutant SOD1G93A astrocytes were previously shown to be toxic to wild-type motor neurons [72, 214]. Interestingly, pretreatment of mutant SOD1G93A astrocytes with low doses of MitoQ reduced oxidative damage and enhanced mitochondrial ATP generation in motor neurons [29]. Adding MitoQ to the drinking water of mutant SOD1G93A mice slowed the decline of mitochondrial function in spinal cord and muscle, reduced spinal cord oxidative damage, improved the integrity of neuromuscular junctions, increased hindlimb strength, and prolonged the life span of mutant SOD1G93A mice [132]. While these results are promising, no clinical trials assessed the efficacy or tolerability of MitoQ in ALS patients thus far.

Dexpramipexole

Pramipexole is a dopamine agonist approved to treat Parkinson’s disease [171] and restless leg syndrome [125]. In addition, pramipexole demonstrates anti-oxidative properties [107]. Dexpramipexole, the R+ enantiomer of pramipexole, has a 100-fold lower affinity for dopamine receptors than pramipexole, but is equipotent to scavenge ROS [44, 68]. Dexpramipexole improved metabolic efficiency, defined as the amount of ATP generated for a given value of oxygen consumption, in whole rat brain-derived mitochondria [2]. In mutant SOD1G93A mice, dexpramipexole prolonged survival and delayed motor deterioration [44]. In a phase II clinical trial, dexpramipexole administration to ALS patients was well tolerated and tended to attenuate functional decline in a dose-dependent manner [39]. However, dexpramipexole did not differ from placebo for any efficacy measurement in a subsequent phase III clinical trial [40]. Moreover, dexpramipexole did not show a protective effect in subsequent preclinical studies in mutant SOD1G93A mice [202], ALS patient derived iPSCs [216], or rat cortical neurons transfected with mutant or wild-type human TDP-43 [202].

Fueling energy metabolism

Imbalanced energy homeostasis is an early and persistent observation throughout the course of ALS. Moreover, endogenous energy stores, which are mainly located in skeletal muscle and adipose tissue, are progressively depleted during disease progression. Providing additional energetic substrates may, therefore, improve the clinical outcome. In addition, impairments in carbohydrate metabolism suggest that energy substrates other than glucose might have more pronounced effects. Enhancing the availability of specific metabolites is an alternative way to improve ATP production. This could be particularly important in neurons to compensate for losses of the TCA intermediate α-ketoglutarate that occur through the release of the α-ketoglutarate-derived neurotransmitters glutamate and GABA [169].

High caloric diets to treat ALS

In mutant SOD1G93A, SOD1G86R, and TDP43A315T overexpressing mice [36, 56], a high fat diet delayed disease onset and extended survival, while caloric restriction shortened the lifespan of mutant SOD1G93A mice [145, 146]. Moreover, a high fat diet attenuated motor neuron loss in the spinal cord of mutant SOD1G93A mice [56]. A small prospective study in ALS patients showed that high caloric diets were able to abolish weight loss [51]. In a phase II clinical trial involving 20 patients, subjects were assigned to one of three diets using gastrostomy: caloric intake designed to match caloric expenditure, a high fat diet, or a high carbohydrate diet both providing an excess of calories. One out of 17 patients assigned to a hypercaloric diet died during the 5-month follow-up period compared with three out of seven patients assigned to the control group [208]. While this study was promising, a sufficiently powered phase III clinical trial to examine the effect of hypercaloric diets on survival and functional outcome in ALS patients is still lacking. Moreover, whether the composition of the hypercaloric diet matters is an outstanding question.

Ketone bodies, ketogenic diets, and beyond

Ketone bodies are energy substrates endogenously produced from fat when glucose availability is limited. While the liver is the major site of ketogenesis, glial cells are also able to produce ketone bodies [5]. Ketone bodies have a high metabolic efficiency generating 30% more energy per molecule oxygen than pyruvate [199]. Therefore, ketones are suited to meet high-energy demands. Besides being an energy substrate, ketone bodies are signaling metabolites acting as histone deacetylase inhibitors to reduce oxidative stress [177]. In mutant SOD1G93A mice, a ketogenic diet delayed disease onset and improved motor neuron survival without affecting lifespan [222]. However, as ketogenic diets are associated with a loss of muscle mass [24], the potential beneficial effect of ketosis on lifespan may be blunted. Recently, ketone esters have emerged as a novel approach to raise blood ketone bodies immediately [34, 97], even when co-ingesting high amounts of carbohydrates and proteins [198]. Ketone esters have improved disease outcome in an Alzheimer’s disease mouse model [93]. In ALS, the therapeutic potential of ketone esters is unexplored.

Medium-chain triglycerides

Medium-chain triglycerides were previously used as a more palatable alternative to ketogenic diets to treat epilepsy and Alzheimer’s disease [181]. Medium-chain triglycerides are able to cross the blood–brain barrier via diffusion and enter neurons via monocarboxylate transporters [168] where they can undergo β-oxidation to form ACoA and ketone bodies, which fuel the TCA cycle [192]. Two medium-chain triglycerides investigated in the context of ALS are caprylic triglyceride and triheptanoin, the triglycerides of octanoic acid (8C fatty acid) and heptanoic acid (7C fatty acid), respectively. Early administration of caprylic triglyceride to mutant SOD1G93A mice delayed disease onset, improved motor performance, reduced motor neuron loss, and promoted mitochondrial oxygen consumption in the spinal cord [220]. Pre-symptomatic triheptanoin ingestion also delayed disease onset and reduced motor neuron loss at symptom onset in mutant SOD1G93A mice [191]. Clinical trials evaluating safety or efficacy of medium-chain triglyceride treatments in ALS patients are lacking.

Pyruvate

Pyruvate is the end product of glycolysis and represents a mitochondrial fuel entering the TCA cycle after conversion to ACoA. Pyruvate is neuroprotective in models for epilepsy [153] and Alzheimer’s disease [83]. The neuroprotective properties of pyruvate are multifaceted and originate from anti-oxidant properties, the ability to facilitate glutamate efflux from the brain, anti-inflammatory effects, and their ability to increase TCA cycling [223]. In the context of ALS, administration of 1 g pyruvate/kg body weight/week to mutant SOD1G93A mice prolonged the lifespan by 12 days, slowed disease progression, and improved motor performance when starting the treatment at the age of 70 days [142]. However, another study in which mutant SOD1G93A mice received 0.5 g/kg body weight six times a week starting from the same age did not improve survival or rotarod performance [59]. There are no clinical trials available assessing the effect of pyruvate intake in ALS patients.

Enhancing mitochondrial function

In the presence of decreased mitochondrial function, providing additional and/or alternative energy substrates may be insufficient. Enhancing mitochondrial function and hence metabolic efficiency may be necessary.

Dichloroacetate

Increasing the conversion of pyruvate into ACoA, by providing the pyruvate dehydrogenase kinase inhibitor dichloroacetate improved survival, delayed disease onset, and reduced spinal motor neuron loss in mutant SOD1G93A mice [131, 141]. As dichloroacetate blunted the reduction of expression of mitochondrial genes seen in mutant SOD1G93A skeletal muscle during disease progression, improving mitochondrial function could elicit this effect [141]. While in NSC34 motor neuron-like cells mutant SOD1 increased pyruvate dehydrogenase kinase expression and lactate production [195], it was not tested if dichloroacetate treatment improved mitochondrial function in the CNS of mutant SOD1G93A mice. Although doses of dichloroacetate used in preclinical ALS studies are well tolerated in patients with advanced solid tumors [33], ALS clinical trials are lacking.

Acetyl-l-carnitine

Acetyl-l-carnitine is the acetyl-ester of l-carnitine. Acetyl-l-carnitine is an important cellular source of acetyl groups to generate ACoA in high-energy demanding situations [139]. Acetyl-l-carnitine also mediates transport of long chain fatty acids across mitochondrial membranes and is, therefore, rate limiting for β-oxidation. While the neuroprotective effects of acetyl-l-carnitine are mainly described in cortical neurons [166, 175], an early study showed neuroprotective and neurotrophic effects of acetyl-l-carnitine in embryonic rat motor neurons [14]. Moreover, administration of l-carnitine to symptomatic mutant SOD1G93A mice improved survival [100]. Based on these findings, a randomized double-blind placebo-controlled phase II trial was performed in 82 patients. Subjects ingested 3 g of acetyl-l-carnitine or placebo each day together with riluzole. Acetyl-l-carnitine was well tolerated, and respiratory capacity and ALSFRS showed mild improvements. In addition, median survival doubled in the acetyl-l-carnitine group compared to the placebo group [11]. Despite these results, a larger phase III trial has not yet been performed.

Conclusion and future perspectives

While dysregulated systemic energy metabolism is now well established in ALS patients, energy metabolism has received a little attention in ALS research due to its association with mutant SOD1 models. It now becomes obvious that abnormal energy metabolism also has a role in more recently developed ALS models [118, 170, 183, 205]. In ALS motor neurons and glia, both mitochondrial and glycolytic energy metabolism seem to be impaired, but the molecular mechanisms underlying energetic stress remain unknown. Since motor neuron physiology is highly energy demanding, impairments in energy metabolism could, at least in part, explain the selective dying of motor neurons in ALS. As a consequence, targeting defects in energy metabolism in ALS represents a rational therapeutic strategy. Manipulating energy metabolism is a particularly potent strategy to treat complex diseases due to its intimate link to epigenetic control [60, 94, 119] and is, therefore, increasingly recognized as therapeutic target in cancer [28], immunodeficiency [123], and stroke [156]. To date, a unifying view on how different metabolic pathways converge and whether metabolic alterations contribute to disease etiology in ALS is non-existing. Future work using direct measurements of metabolic fluxes is clearly needed to obtain a more in-depth understanding of motor neuron metabolism in health and disease. Moreover, due to the compartmentalization of specific energy requiring processes in motor neurons, defining the role of metabolism and ALS-related motor neuron dysfunction requires high-resolution and spatial subdivision of metabolic and functional analyses. Knowing how ALS motor neurons differ metabolically from healthy motor neurons could offer the necessary insights to develop future therapeutic approaches in ALS. Another relevant area for future research is to explore the metabolic crosstalk between motor neurons and glial cells, as well as other disease-relevant cells such as the muscle.
In conclusion, it is too early to consider ALS at present as a metabolic disease. However, the massive amount circumstantial evidence linking energy metabolism with ALS pathophysiology underscores the therapeutic potential of targeting metabolism. As is the case for many other pathways and mechanisms proposed to play a crucial role in ALS, the ultimate proof that disturbances in metabolism are causally linked to the selective motor neuron death in ALS will be a positive clinical trial with a therapeutic strategy tackling energy metabolism in patients. In the meantime, we strongly believe that a better understanding of the metabolic biology of ALS could lead to the identification of novel therapeutic targets.

Acknowledgements

Work of the authors is supported by Grants from the KU Leuven (C1 and “Opening the Future” Fund), VIB, the “Fund for Scientific Research Flanders” (FWO-Vlaanderen), the Belgian government (Interuniversity Attraction Poles Programme P7/16 initiated by the Belgian Federal Science Policy Office), the Thierry Latran Foundation, the Association Belge contre les Maladies neuro-Musculaires (ABMM), and the ALS Liga Belgium (“A Cure for ALS”). TV is supported by a strategic basic research Ph.D. Grant awarded by the FWO (1S60116N).

Compliance with ethical standards

Conflict of interest

The authors declare no conflict of interest.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

e.Med Neurologie

Kombi-Abonnement

Mit e.Med Neurologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes, den Premium-Inhalten der neurologischen Fachzeitschriften, inklusive einer gedruckten Neurologie-Zeitschrift Ihrer Wahl.

Literatur
5.
Zurück zum Zitat Auestad N, Korsak RA, Morrow JW, Edmond J (1991) Fatty acid oxidation and ketogenesis by astrocytes in primary culture. J Neurochem 56:1376–1386PubMedCrossRef Auestad N, Korsak RA, Morrow JW, Edmond J (1991) Fatty acid oxidation and ketogenesis by astrocytes in primary culture. J Neurochem 56:1376–1386PubMedCrossRef
10.
Zurück zum Zitat Bayer H, Lang K, Buck E, Higelin J, Barteczko L, Pasquarelli N, Sprissler J, Lucas T, Holzmann K, Demestre M, Lindenberg KS, Danzer KM, Boeckers T, Ludolph AC, Dupuis L, Weydt P, Witting A (2017) ALS-causing mutations differentially affect PGC-1alpha expression and function in the brain vs. peripheral tissues. Neurobiol Dis 97:36–45. https://doi.org/10.1016/j.nbd.2016.11.001 PubMedCrossRef Bayer H, Lang K, Buck E, Higelin J, Barteczko L, Pasquarelli N, Sprissler J, Lucas T, Holzmann K, Demestre M, Lindenberg KS, Danzer KM, Boeckers T, Ludolph AC, Dupuis L, Weydt P, Witting A (2017) ALS-causing mutations differentially affect PGC-1alpha expression and function in the brain vs. peripheral tissues. Neurobiol Dis 97:36–45. https://​doi.​org/​10.​1016/​j.​nbd.​2016.​11.​001 PubMedCrossRef
11.
Zurück zum Zitat Beghi E, Pupillo E, Bonito V, Buzzi P, Caponnetto C, Chio A, Corbo M, Giannini F, Inghilleri M, Bella VL, Logroscino G, Lorusso L, Lunetta C, Mazzini L, Messina P, Mora G, Perini M, Quadrelli ML, Silani V, Simone IL, Tremolizzo L, Italian ALSSG (2013) Randomized double-blind placebo-controlled trial of acetyl-l-carnitine for ALS. Amyotroph Lateral Scler Frontotemporal Degener 14:397–405. https://doi.org/10.3109/21678421.2013.764568 PubMedCrossRef Beghi E, Pupillo E, Bonito V, Buzzi P, Caponnetto C, Chio A, Corbo M, Giannini F, Inghilleri M, Bella VL, Logroscino G, Lorusso L, Lunetta C, Mazzini L, Messina P, Mora G, Perini M, Quadrelli ML, Silani V, Simone IL, Tremolizzo L, Italian ALSSG (2013) Randomized double-blind placebo-controlled trial of acetyl-l-carnitine for ALS. Amyotroph Lateral Scler Frontotemporal Degener 14:397–405. https://​doi.​org/​10.​3109/​21678421.​2013.​764568 PubMedCrossRef
14.
Zurück zum Zitat Bigini P, Larini S, Pasquali C, Muzio V, Mennini T (2002) Acetyl-l-carnitine shows neuroprotective and neurotrophic activity in primary culture of rat embryo motoneurons. Neurosci Lett 329:334–338PubMedCrossRef Bigini P, Larini S, Pasquali C, Muzio V, Mennini T (2002) Acetyl-l-carnitine shows neuroprotective and neurotrophic activity in primary culture of rat embryo motoneurons. Neurosci Lett 329:334–338PubMedCrossRef
16.
Zurück zum Zitat Boero J, Qin W, Cheng J, Woolsey TA, Strauss AW, Khuchua Z (2003) Restricted neuronal expression of ubiquitous mitochondrial creatine kinase: changing patterns in development and with increased activity. Mol Cell Biochem 244:69–76PubMedCrossRef Boero J, Qin W, Cheng J, Woolsey TA, Strauss AW, Khuchua Z (2003) Restricted neuronal expression of ubiquitous mitochondrial creatine kinase: changing patterns in development and with increased activity. Mol Cell Biochem 244:69–76PubMedCrossRef
19.
Zurück zum Zitat Borthwick GM, Johnson MA, Ince PG, Shaw PJ, Turnbull DM (1999) Mitochondrial enzyme activity in amyotrophic lateral sclerosis: implications for the role of mitochondria in neuronal cell death. Ann Neurol 46:787–790PubMedCrossRef Borthwick GM, Johnson MA, Ince PG, Shaw PJ, Turnbull DM (1999) Mitochondrial enzyme activity in amyotrophic lateral sclerosis: implications for the role of mitochondria in neuronal cell death. Ann Neurol 46:787–790PubMedCrossRef
23.
Zurück zum Zitat Bowling AC, Schulz JB, Brown RH Jr, Beal MF (1993) Superoxide dismutase activity, oxidative damage, and mitochondrial energy metabolism in familial and sporadic amyotrophic lateral sclerosis. J Neurochem 61:2322–2325PubMedCrossRef Bowling AC, Schulz JB, Brown RH Jr, Beal MF (1993) Superoxide dismutase activity, oxidative damage, and mitochondrial energy metabolism in familial and sporadic amyotrophic lateral sclerosis. J Neurochem 61:2322–2325PubMedCrossRef
27.
Zurück zum Zitat Cai B, Zhu Y, Ma Y, Xu Z, Zao Y, Wang J, Lin Y, Comer GM (2003) Effect of supplementing a high-fat, low-carbohydrate enteral formula in COPD patients. Nutrition 19:229–232PubMedCrossRef Cai B, Zhu Y, Ma Y, Xu Z, Zao Y, Wang J, Lin Y, Comer GM (2003) Effect of supplementing a high-fat, low-carbohydrate enteral formula in COPD patients. Nutrition 19:229–232PubMedCrossRef
28.
Zurück zum Zitat Cantelmo AR, Conradi LC, Brajic A, Goveia J, Kalucka J, Pircher A, Chaturvedi P, Hol J, Thienpont B, Teuwen LA, Schoors S, Boeckx B, Vriens J, Kuchnio A, Veys K, Cruys B, Finotto L, Treps L, Stav-Noraas TE, Bifari F, Stapor P, Decimo I, Kampen K, De Bock K, Haraldsen G, Schoonjans L, Rabelink T, Eelen G, Ghesquiere B, Rehman J, Lambrechts D, Malik AB, Dewerchin M, Carmeliet P (2016) Inhibition of the glycolytic activator PFKFB3 in endothelium induces tumor vessel normalization, impairs metastasis, and improves chemotherapy. Cancer Cell 30:968–985. https://doi.org/10.1016/j.ccell.2016.10.006 PubMedPubMedCentralCrossRef Cantelmo AR, Conradi LC, Brajic A, Goveia J, Kalucka J, Pircher A, Chaturvedi P, Hol J, Thienpont B, Teuwen LA, Schoors S, Boeckx B, Vriens J, Kuchnio A, Veys K, Cruys B, Finotto L, Treps L, Stav-Noraas TE, Bifari F, Stapor P, Decimo I, Kampen K, De Bock K, Haraldsen G, Schoonjans L, Rabelink T, Eelen G, Ghesquiere B, Rehman J, Lambrechts D, Malik AB, Dewerchin M, Carmeliet P (2016) Inhibition of the glycolytic activator PFKFB3 in endothelium induces tumor vessel normalization, impairs metastasis, and improves chemotherapy. Cancer Cell 30:968–985. https://​doi.​org/​10.​1016/​j.​ccell.​2016.​10.​006 PubMedPubMedCentralCrossRef
37.
38.
Zurück zum Zitat Crugnola V, Lamperti C, Lucchini V, Ronchi D, Peverelli L, Prelle A, Sciacco M, Bordoni A, Fassone E, Fortunato F, Corti S, Silani V, Bresolin N, Di Mauro S, Comi GP, Moggio M (2010) Mitochondrial respiratory chain dysfunction in muscle from patients with amyotrophic lateral sclerosis. Arch Neurol 67:849–854. https://doi.org/10.1001/archneurol.2010.128 PubMedCrossRef Crugnola V, Lamperti C, Lucchini V, Ronchi D, Peverelli L, Prelle A, Sciacco M, Bordoni A, Fassone E, Fortunato F, Corti S, Silani V, Bresolin N, Di Mauro S, Comi GP, Moggio M (2010) Mitochondrial respiratory chain dysfunction in muscle from patients with amyotrophic lateral sclerosis. Arch Neurol 67:849–854. https://​doi.​org/​10.​1001/​archneurol.​2010.​128 PubMedCrossRef
39.
Zurück zum Zitat Cudkowicz M, Bozik ME, Ingersoll EW, Miller R, Mitsumoto H, Shefner J, Moore DH, Schoenfeld D, Mather JL, Archibald D, Sullivan M, Amburgey C, Moritz J, Gribkoff VK (2011) The effects of dexpramipexole (KNS-760704) in individuals with amyotrophic lateral sclerosis. Nat Med 17:1652–1656. https://doi.org/10.1038/nm.2579 PubMedCrossRef Cudkowicz M, Bozik ME, Ingersoll EW, Miller R, Mitsumoto H, Shefner J, Moore DH, Schoenfeld D, Mather JL, Archibald D, Sullivan M, Amburgey C, Moritz J, Gribkoff VK (2011) The effects of dexpramipexole (KNS-760704) in individuals with amyotrophic lateral sclerosis. Nat Med 17:1652–1656. https://​doi.​org/​10.​1038/​nm.​2579 PubMedCrossRef
40.
Zurück zum Zitat Cudkowicz ME, van den Berg LH, Shefner JM, Mitsumoto H, Mora JS, Ludolph A, Hardiman O, Bozik ME, Ingersoll EW, Archibald D, Meyers AL, Dong Y, Farwell WR, Kerr DA, Investigators E (2013) Dexpramipexole versus placebo for patients with amyotrophic lateral sclerosis (EMPOWER): a randomised, double-blind, phase 3 trial. Lancet Neurol 12:1059–1067. https://doi.org/10.1016/S1474-4422(13)70221-7 PubMedCrossRef Cudkowicz ME, van den Berg LH, Shefner JM, Mitsumoto H, Mora JS, Ludolph A, Hardiman O, Bozik ME, Ingersoll EW, Archibald D, Meyers AL, Dong Y, Farwell WR, Kerr DA, Investigators E (2013) Dexpramipexole versus placebo for patients with amyotrophic lateral sclerosis (EMPOWER): a randomised, double-blind, phase 3 trial. Lancet Neurol 12:1059–1067. https://​doi.​org/​10.​1016/​S1474-4422(13)70221-7 PubMedCrossRef
41.
Zurück zum Zitat Dal Canto MC, Gurney ME (1994) Development of central nervous system pathology in a murine transgenic model of human amyotrophic lateral sclerosis. Am J Pathol 145:1271–1279 Dal Canto MC, Gurney ME (1994) Development of central nervous system pathology in a murine transgenic model of human amyotrophic lateral sclerosis. Am J Pathol 145:1271–1279
44.
Zurück zum Zitat Danzeisen R, Schwalenstoecker B, Gillardon F, Buerger E, Krzykalla V, Klinder K, Schild L, Hengerer B, Ludolph AC, Dorner-Ciossek C, Kussmaul L (2006) Targeted antioxidative and neuroprotective properties of the dopamine agonist pramipexole and its nondopaminergic enantiomer SND919CL2x [(+)2-amino-4,5,6,7-tetrahydro-6-Lpropylamino-benzathiazole dihydrochloride]. J Pharmacol Exp Ther 316:189–199. https://doi.org/10.1124/jpet.105.092312 PubMedCrossRef Danzeisen R, Schwalenstoecker B, Gillardon F, Buerger E, Krzykalla V, Klinder K, Schild L, Hengerer B, Ludolph AC, Dorner-Ciossek C, Kussmaul L (2006) Targeted antioxidative and neuroprotective properties of the dopamine agonist pramipexole and its nondopaminergic enantiomer SND919CL2x [(+)2-amino-4,5,6,7-tetrahydro-6-Lpropylamino-benzathiazole dihydrochloride]. J Pharmacol Exp Ther 316:189–199. https://​doi.​org/​10.​1124/​jpet.​105.​092312 PubMedCrossRef
45.
Zurück zum Zitat De Vos KJ, Chapman AL, Tennant ME, Manser C, Tudor EL, Lau KF, Brownlees J, Ackerley S, Shaw PJ, McLoughlin DM, Shaw CE, Leigh PN, Miller CCJ, Grierson AJ (2007) Familial amyotrophic lateral sclerosis-linked SOD1 mutants perturb fast axonal transport to reduce axonal mitochondria content. Hum Mol Genet 16:2720–2728. https://doi.org/10.1093/hmg/ddm226 PubMedPubMedCentralCrossRef De Vos KJ, Chapman AL, Tennant ME, Manser C, Tudor EL, Lau KF, Brownlees J, Ackerley S, Shaw PJ, McLoughlin DM, Shaw CE, Leigh PN, Miller CCJ, Grierson AJ (2007) Familial amyotrophic lateral sclerosis-linked SOD1 mutants perturb fast axonal transport to reduce axonal mitochondria content. Hum Mol Genet 16:2720–2728. https://​doi.​org/​10.​1093/​hmg/​ddm226 PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Derave W, Van Den Bosch L, Lemmens G, Eijnde BO, Robberecht W, Hespel P (2003) Skeletal muscle properties in a transgenic mouse model for amyotrophic lateral sclerosis: effects of creatine treatment. Neurobiol Dis 13:264–272PubMedCrossRef Derave W, Van Den Bosch L, Lemmens G, Eijnde BO, Robberecht W, Hespel P (2003) Skeletal muscle properties in a transgenic mouse model for amyotrophic lateral sclerosis: effects of creatine treatment. Neurobiol Dis 13:264–272PubMedCrossRef
63.
Zurück zum Zitat Ferrante RJ, Browne SE, Shinobu LA, Bowling AC, Baik MJ, MacGarvey U, Kowall NW, Brown RH Jr, Beal MF (1997) Evidence of increased oxidative damage in both sporadic and familial amyotrophic lateral sclerosis. J Neurochem 69:2064–2074PubMedCrossRef Ferrante RJ, Browne SE, Shinobu LA, Bowling AC, Baik MJ, MacGarvey U, Kowall NW, Brown RH Jr, Beal MF (1997) Evidence of increased oxidative damage in both sporadic and familial amyotrophic lateral sclerosis. J Neurochem 69:2064–2074PubMedCrossRef
64.
Zurück zum Zitat Funfschilling U, Supplie LM, Mahad D, Boretius S, Saab AS, Edgar J, Brinkmann BG, Kassmann CM, Tzvetanova ID, Mobius W, Diaz F, Meijer D, Suter U, Hamprecht B, Sereda MW, Moraes CT, Frahm J, Goebbels S, Nave KA (2012) Glycolytic oligodendrocytes maintain myelin and long-term axonal integrity. Nature 485:517–521. https://doi.org/10.1038/nature11007 PubMedPubMedCentralCrossRef Funfschilling U, Supplie LM, Mahad D, Boretius S, Saab AS, Edgar J, Brinkmann BG, Kassmann CM, Tzvetanova ID, Mobius W, Diaz F, Meijer D, Suter U, Hamprecht B, Sereda MW, Moraes CT, Frahm J, Goebbels S, Nave KA (2012) Glycolytic oligodendrocytes maintain myelin and long-term axonal integrity. Nature 485:517–521. https://​doi.​org/​10.​1038/​nature11007 PubMedPubMedCentralCrossRef
65.
Zurück zum Zitat Gallo V, Wark PA, Jenab M, Pearce N, Brayne C, Vermeulen R, Andersen PM, Hallmans G, Kyrozis A, Vanacore N, Vahdaninia M, Grote V, Kaaks R, Mattiello A, Bueno-de-Mesquita HB, Peeters PH, Travis RC, Petersson J, Hansson O, Arriola L, Jimenez-Martin JM, Tjonneland A, Halkjaer J, Agnoli C, Sacerdote C, Bonet C, Trichopoulou A, Gavrila D, Overvad K, Weiderpass E, Palli D, Quiros JR, Tumino R, Khaw KT, Wareham N, Barricante-Gurrea A, Fedirko V, Ferrari P, Clavel-Chapelon F, Boutron-Ruault MC, Boeing H, Vigl M, Middleton L, Riboli E, Vineis P (2013) Prediagnostic body fat and risk of death from amyotrophic lateral sclerosis: the EPIC cohort. Neurology 80:829–838. https://doi.org/10.1212/WNL.0b013e3182840689 PubMedPubMedCentralCrossRef Gallo V, Wark PA, Jenab M, Pearce N, Brayne C, Vermeulen R, Andersen PM, Hallmans G, Kyrozis A, Vanacore N, Vahdaninia M, Grote V, Kaaks R, Mattiello A, Bueno-de-Mesquita HB, Peeters PH, Travis RC, Petersson J, Hansson O, Arriola L, Jimenez-Martin JM, Tjonneland A, Halkjaer J, Agnoli C, Sacerdote C, Bonet C, Trichopoulou A, Gavrila D, Overvad K, Weiderpass E, Palli D, Quiros JR, Tumino R, Khaw KT, Wareham N, Barricante-Gurrea A, Fedirko V, Ferrari P, Clavel-Chapelon F, Boutron-Ruault MC, Boeing H, Vigl M, Middleton L, Riboli E, Vineis P (2013) Prediagnostic body fat and risk of death from amyotrophic lateral sclerosis: the EPIC cohort. Neurology 80:829–838. https://​doi.​org/​10.​1212/​WNL.​0b013e3182840689​ PubMedPubMedCentralCrossRef
67.
70.
Zurück zum Zitat Guo W, Naujock M, Fumagalli L, Vandoorne T, Baatsen P, Boon R, Ordovas L, Patel A, Welters M, Vanwelden T, Geens N, Tricot T, Benoy V, Steyaert J, Lefebvre-Omar C, Boesmans W, Jarpe M, Sterneckert J, Wegner F, Petri S, Bohl D, Vanden Berghe P, Robberecht W, Van Damme P, Verfaillie C, Van Den Bosch L (2017) HDAC6 inhibition reverses axonal transport defects in motor neurons derived from FUS-ALS patients. Nat Commun 8:861. https://doi.org/10.1038/s41467-017-00911-y PubMedPubMedCentralCrossRef Guo W, Naujock M, Fumagalli L, Vandoorne T, Baatsen P, Boon R, Ordovas L, Patel A, Welters M, Vanwelden T, Geens N, Tricot T, Benoy V, Steyaert J, Lefebvre-Omar C, Boesmans W, Jarpe M, Sterneckert J, Wegner F, Petri S, Bohl D, Vanden Berghe P, Robberecht W, Van Damme P, Verfaillie C, Van Den Bosch L (2017) HDAC6 inhibition reverses axonal transport defects in motor neurons derived from FUS-ALS patients. Nat Commun 8:861. https://​doi.​org/​10.​1038/​s41467-017-00911-y PubMedPubMedCentralCrossRef
72.
76.
Zurück zum Zitat Hasel P, Dando O, Jiwaji Z, Baxter P, Todd AC, Heron S, Markus NM, McQueen J, Hampton DW, Torvell M, Tiwari SS, McKay S, Eraso-Pichot A, Zorzano A, Masgrau R, Galea E, Chandran S, Wyllie DJA, Simpson TI, Hardingham GE (2017) Neurons and neuronal activity control gene expression in astrocytes to regulate their development and metabolism. Nat Commun 8:15132. https://doi.org/10.1038/ncomms15132 PubMedPubMedCentralCrossRef Hasel P, Dando O, Jiwaji Z, Baxter P, Todd AC, Heron S, Markus NM, McQueen J, Hampton DW, Torvell M, Tiwari SS, McKay S, Eraso-Pichot A, Zorzano A, Masgrau R, Galea E, Chandran S, Wyllie DJA, Simpson TI, Hardingham GE (2017) Neurons and neuronal activity control gene expression in astrocytes to regulate their development and metabolism. Nat Commun 8:15132. https://​doi.​org/​10.​1038/​ncomms15132 PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Hatazawa J, Brooks RA, Dalakas MC, Mansi L, Di Chiro G (1988) Cortical motor-sensory hypometabolism in amyotrophic lateral sclerosis: a PET study. J Comput Assist Tomogr 12:630–636PubMedCrossRef Hatazawa J, Brooks RA, Dalakas MC, Mansi L, Di Chiro G (1988) Cortical motor-sensory hypometabolism in amyotrophic lateral sclerosis: a PET study. J Comput Assist Tomogr 12:630–636PubMedCrossRef
78.
83.
Zurück zum Zitat Isopi E, Granzotto A, Corona C, Bomba M, Ciavardelli D, Curcio M, Canzoniero LM, Navarra R, Lattanzio R, Piantelli M, Sensi SL (2015) Pyruvate prevents the development of age-dependent cognitive deficits in a mouse model of Alzheimer’s disease without reducing amyloid and tau pathology. Neurobiol Dis 81:214–224. https://doi.org/10.1016/j.nbd.2014.11.013 PubMedCrossRef Isopi E, Granzotto A, Corona C, Bomba M, Ciavardelli D, Curcio M, Canzoniero LM, Navarra R, Lattanzio R, Piantelli M, Sensi SL (2015) Pyruvate prevents the development of age-dependent cognitive deficits in a mouse model of Alzheimer’s disease without reducing amyloid and tau pathology. Neurobiol Dis 81:214–224. https://​doi.​org/​10.​1016/​j.​nbd.​2014.​11.​013 PubMedCrossRef
84.
Zurück zum Zitat Jaarsma D, Haasdijk ED, Grashorn JA, Hawkins R, van Duijn W, Verspaget HW, London J, Holstege JC (2000) Human Cu/Zn superoxide dismutase (SOD1) overexpression in mice causes mitochondrial vacuolization, axonal degeneration, and premature motoneuron death and accelerates motoneuron disease in mice expressing a familial amyotrophic lateral sclerosis mutant SOD1. Neurobiol Dis 7:623–643. https://doi.org/10.1006/nbdi.2000.0299 PubMedCrossRef Jaarsma D, Haasdijk ED, Grashorn JA, Hawkins R, van Duijn W, Verspaget HW, London J, Holstege JC (2000) Human Cu/Zn superoxide dismutase (SOD1) overexpression in mice causes mitochondrial vacuolization, axonal degeneration, and premature motoneuron death and accelerates motoneuron disease in mice expressing a familial amyotrophic lateral sclerosis mutant SOD1. Neurobiol Dis 7:623–643. https://​doi.​org/​10.​1006/​nbdi.​2000.​0299 PubMedCrossRef
90.
Zurück zum Zitat Jung C, Higgins CM, Xu Z (2002) Mitochondrial electron transport chain complex dysfunction in a transgenic mouse model for amyotrophic lateral sclerosis. J Neurochem 83:535–545PubMedCrossRef Jung C, Higgins CM, Xu Z (2002) Mitochondrial electron transport chain complex dysfunction in a transgenic mouse model for amyotrophic lateral sclerosis. J Neurochem 83:535–545PubMedCrossRef
95.
Zurück zum Zitat Kaufmann P, Thompson JL, Levy G, Buchsbaum R, Shefner J, Krivickas LS, Katz J, Rollins Y, Barohn RJ, Jackson CE, Tiryaki E, Lomen-Hoerth C, Armon C, Tandan R, Rudnicki SA, Rezania K, Sufit R, Pestronk A, Novella SP, Heiman-Patterson T, Kasarskis EJ, Pioro EP, Montes J, Arbing R, Vecchio D, Barsdorf A, Mitsumoto H, Levin B, Group QS (2009) Phase II trial of CoQ10 for ALS finds insufficient evidence to justify phase III. Ann Neurol 66:235–244. https://doi.org/10.1002/ana.21743 PubMedPubMedCentralCrossRef Kaufmann P, Thompson JL, Levy G, Buchsbaum R, Shefner J, Krivickas LS, Katz J, Rollins Y, Barohn RJ, Jackson CE, Tiryaki E, Lomen-Hoerth C, Armon C, Tandan R, Rudnicki SA, Rezania K, Sufit R, Pestronk A, Novella SP, Heiman-Patterson T, Kasarskis EJ, Pioro EP, Montes J, Arbing R, Vecchio D, Barsdorf A, Mitsumoto H, Levin B, Group QS (2009) Phase II trial of CoQ10 for ALS finds insufficient evidence to justify phase III. Ann Neurol 66:235–244. https://​doi.​org/​10.​1002/​ana.​21743 PubMedPubMedCentralCrossRef
97.
Zurück zum Zitat Kesl SL, Poff AM, Ward NP, Fiorelli TN, Ari C, Van Putten AJ, Sherwood JW, Arnold P, D’Agostino DP (2016) Effects of exogenous ketone supplementation on blood ketone, glucose, triglyceride, and lipoprotein levels in Sprague–Dawley rats. Nutr Metab (Lond) 13:9. https://doi.org/10.1186/s12986-016-0069-y CrossRef Kesl SL, Poff AM, Ward NP, Fiorelli TN, Ari C, Van Putten AJ, Sherwood JW, Arnold P, D’Agostino DP (2016) Effects of exogenous ketone supplementation on blood ketone, glucose, triglyceride, and lipoprotein levels in Sprague–Dawley rats. Nutr Metab (Lond) 13:9. https://​doi.​org/​10.​1186/​s12986-016-0069-y CrossRef
101.
Zurück zum Zitat Klivenyi P, Ferrante RJ, Matthews RT, Bogdanov MB, Klein AM, Andreassen OA, Mueller G, Wermer M, Kaddurah-Daouk R, Beal MF (1999) Neuroprotective effects of creatine in a transgenic animal model of amyotrophic lateral sclerosis. Nat Med 5:347–350. https://doi.org/10.1038/6568 PubMedCrossRef Klivenyi P, Ferrante RJ, Matthews RT, Bogdanov MB, Klein AM, Andreassen OA, Mueller G, Wermer M, Kaddurah-Daouk R, Beal MF (1999) Neuroprotective effects of creatine in a transgenic animal model of amyotrophic lateral sclerosis. Nat Med 5:347–350. https://​doi.​org/​10.​1038/​6568 PubMedCrossRef
102.
Zurück zum Zitat Kong JM, Xu ZS (1998) Massive mitochondrial degeneration in motor neurons triggers the onset of amyotrophic lateral sclerosis in mice expressing a mutant SOD1. J Neurosci 18:3241–3250PubMed Kong JM, Xu ZS (1998) Massive mitochondrial degeneration in motor neurons triggers the onset of amyotrophic lateral sclerosis in mice expressing a mutant SOD1. J Neurosci 18:3241–3250PubMed
103.
Zurück zum Zitat Lacomblez L, Doppler V, Beucler I, Costes G, Salachas F, Raisonnier A, Le Forestier N, Pradat PF, Bruckert E, Meininger V (2002) APOE: a potential marker of disease progression in ALS. Neurology 58:1112–1114PubMedCrossRef Lacomblez L, Doppler V, Beucler I, Costes G, Salachas F, Raisonnier A, Le Forestier N, Pradat PF, Bruckert E, Meininger V (2002) APOE: a potential marker of disease progression in ALS. Neurology 58:1112–1114PubMedCrossRef
115.
Zurück zum Zitat Llorente-Folch I, Rueda CB, Amigo I, del Arco A, Saheki T, Pardo B, Satrustegui J (2013) Calcium-regulation of mitochondrial respiration maintains ATP homeostasis and requires ARALAR/AGC1-malate aspartate shuttle in intact cortical neurons. J Neurosci 33(13957–13971):13971a. https://doi.org/10.1523/JNEUROSCI.0929-13.2013 Llorente-Folch I, Rueda CB, Amigo I, del Arco A, Saheki T, Pardo B, Satrustegui J (2013) Calcium-regulation of mitochondrial respiration maintains ATP homeostasis and requires ARALAR/AGC1-malate aspartate shuttle in intact cortical neurons. J Neurosci 33(13957–13971):13971a. https://​doi.​org/​10.​1523/​JNEUROSCI.​0929-13.​2013
120.
Zurück zum Zitat Machler P, Wyss MT, Elsayed M, Stobart J, Gutierrez R, von Faber-Castell A, Kaelin V, Zuend M, San Martin A, Romero-Gomez I, Baeza-Lehnert F, Lengacher S, Schneider BL, Aebischer P, Magistretti PJ, Barros LF, Weber B (2016) In vivo evidence for a lactate gradient from astrocytes to neurons. Cell Metab 23:94–102. https://doi.org/10.1016/j.cmet.2015.10.010 PubMedCrossRef Machler P, Wyss MT, Elsayed M, Stobart J, Gutierrez R, von Faber-Castell A, Kaelin V, Zuend M, San Martin A, Romero-Gomez I, Baeza-Lehnert F, Lengacher S, Schneider BL, Aebischer P, Magistretti PJ, Barros LF, Weber B (2016) In vivo evidence for a lactate gradient from astrocytes to neurons. Cell Metab 23:94–102. https://​doi.​org/​10.​1016/​j.​cmet.​2015.​10.​010 PubMedCrossRef
123.
Zurück zum Zitat Mak TW, Grusdat M, Duncan GS, Dostert C, Nonnenmacher Y, Cox M, Binsfeld C, Hao Z, Brustle A, Itsumi M, Jager C, Chen Y, Pinkenburg O, Camara B, Ollert M, Bindslev-Jensen C, Vasiliou V, Gorrini C, Lang PA, Lohoff M, Harris IS, Hiller K, Brenner D (2017) Glutathione primes T cell metabolism for inflammation. Immunity 46:675–689. https://doi.org/10.1016/j.immuni.2017.03.019 PubMedCrossRef Mak TW, Grusdat M, Duncan GS, Dostert C, Nonnenmacher Y, Cox M, Binsfeld C, Hao Z, Brustle A, Itsumi M, Jager C, Chen Y, Pinkenburg O, Camara B, Ollert M, Bindslev-Jensen C, Vasiliou V, Gorrini C, Lang PA, Lohoff M, Harris IS, Hiller K, Brenner D (2017) Glutathione primes T cell metabolism for inflammation. Immunity 46:675–689. https://​doi.​org/​10.​1016/​j.​immuni.​2017.​03.​019 PubMedCrossRef
128.
Zurück zum Zitat Matthews RT, Yang L, Browne S, Baik M, Beal MF (1998) Coenzyme Q10 administration increases brain mitochondrial concentrations and exerts neuroprotective effects. Proc Natl Acad Sci USA 95:8892–8897PubMedPubMedCentralCrossRef Matthews RT, Yang L, Browne S, Baik M, Beal MF (1998) Coenzyme Q10 administration increases brain mitochondrial concentrations and exerts neuroprotective effects. Proc Natl Acad Sci USA 95:8892–8897PubMedPubMedCentralCrossRef
130.
Zurück zum Zitat Menzies FM, Ince PG, Shaw PJ (2002) Mitochondrial involvement in amyotrophic lateral sclerosis. Neurochem Int 40:543–551PubMedCrossRef Menzies FM, Ince PG, Shaw PJ (2002) Mitochondrial involvement in amyotrophic lateral sclerosis. Neurochem Int 40:543–551PubMedCrossRef
136.
Zurück zum Zitat Neumann M, Sampathu DM, Kwong LK, Truax AC, Micsenyi MC, Chou TT, Bruce J, Schuck T, Grossman M, Clark CM, McCluskey LF, Miller BL, Masliah E, Mackenzie IR, Feldman H, Feiden W, Kretzschmar HA, Trojanowski JQ, Lee VM (2006) Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science 314:130–133. https://doi.org/10.1126/science.1134108 PubMedCrossRef Neumann M, Sampathu DM, Kwong LK, Truax AC, Micsenyi MC, Chou TT, Bruce J, Schuck T, Grossman M, Clark CM, McCluskey LF, Miller BL, Masliah E, Mackenzie IR, Feldman H, Feiden W, Kretzschmar HA, Trojanowski JQ, Lee VM (2006) Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science 314:130–133. https://​doi.​org/​10.​1126/​science.​1134108 PubMedCrossRef
146.
Zurück zum Zitat Pedersen WA, Mattson MP (1999) No benefit of dietary restriction on disease onset or progression in amyotrophic lateral sclerosis Cu/Zn-superoxide dismutase mutant mice. Brain Res 833:117–120PubMedCrossRef Pedersen WA, Mattson MP (1999) No benefit of dietary restriction on disease onset or progression in amyotrophic lateral sclerosis Cu/Zn-superoxide dismutase mutant mice. Brain Res 833:117–120PubMedCrossRef
147.
Zurück zum Zitat Pellerin L, Magistretti PJ (1994) Glutamate uptake into astrocytes stimulates aerobic glycolysis: a mechanism coupling neuronal activity to glucose utilization. Proc Natl Acad Sci USA 91:10625–10629PubMedPubMedCentralCrossRef Pellerin L, Magistretti PJ (1994) Glutamate uptake into astrocytes stimulates aerobic glycolysis: a mechanism coupling neuronal activity to glucose utilization. Proc Natl Acad Sci USA 91:10625–10629PubMedPubMedCentralCrossRef
154.
Zurück zum Zitat Pradat PF, Bruneteau G, Gordon PH, Dupuis L, Bonnefont-Rousselot D, Simon D, Salachas F, Corcia P, Frochot V, Lacorte JM, Jardel C, Coussieu C, Le Forestier N, Lacomblez L, Loeffler JP, Meininger V (2010) Impaired glucose tolerance in patients with amyotrophic lateral sclerosis. Amyotroph Lateral Scler 11:166–171. https://doi.org/10.3109/17482960902822960 PubMedCrossRef Pradat PF, Bruneteau G, Gordon PH, Dupuis L, Bonnefont-Rousselot D, Simon D, Salachas F, Corcia P, Frochot V, Lacorte JM, Jardel C, Coussieu C, Le Forestier N, Lacomblez L, Loeffler JP, Meininger V (2010) Impaired glucose tolerance in patients with amyotrophic lateral sclerosis. Amyotroph Lateral Scler 11:166–171. https://​doi.​org/​10.​3109/​1748296090282296​0 PubMedCrossRef
156.
Zurück zum Zitat Quaegebeur A, Segura I, Schmieder R, Verdegem D, Decimo I, Bifari F, Dresselaers T, Eelen G, Ghosh D, Davidson SM, Schoors S, Broekaert D, Cruys B, Govaerts K, De Legher C, Bouche A, Schoonjans L, Ramer MS, Hung G, Bossaert G, Cleveland DW, Himmelreich U, Voets T, Lemmens R, Bennett CF, Robberecht W, De Bock K, Dewerchin M, Ghesquiere B, Fendt SM, Carmeliet P (2016) Deletion or inhibition of the oxygen sensor PHD1 protects against ischemic stroke via reprogramming of neuronal metabolism. Cell Metab 23:280–291. https://doi.org/10.1016/j.cmet.2015.12.007 PubMedPubMedCentralCrossRef Quaegebeur A, Segura I, Schmieder R, Verdegem D, Decimo I, Bifari F, Dresselaers T, Eelen G, Ghosh D, Davidson SM, Schoors S, Broekaert D, Cruys B, Govaerts K, De Legher C, Bouche A, Schoonjans L, Ramer MS, Hung G, Bossaert G, Cleveland DW, Himmelreich U, Voets T, Lemmens R, Bennett CF, Robberecht W, De Bock K, Dewerchin M, Ghesquiere B, Fendt SM, Carmeliet P (2016) Deletion or inhibition of the oxygen sensor PHD1 protects against ischemic stroke via reprogramming of neuronal metabolism. Cell Metab 23:280–291. https://​doi.​org/​10.​1016/​j.​cmet.​2015.​12.​007 PubMedPubMedCentralCrossRef
157.
Zurück zum Zitat Raman R, Allen SP, Goodall EF, Kramer S, Ponger LL, Heath PR, Milo M, Hollinger HC, Walsh T, Highley JR, Olpin S, McDermott CJ, Shaw PJ, Kirby J (2015) Gene expression signatures in motor neurone disease fibroblasts reveal dysregulation of metabolism, hypoxia-response and RNA processing functions. Neuropathol Appl Neurobiol 41:201–226. https://doi.org/10.1111/nan.12147 PubMedPubMedCentralCrossRef Raman R, Allen SP, Goodall EF, Kramer S, Ponger LL, Heath PR, Milo M, Hollinger HC, Walsh T, Highley JR, Olpin S, McDermott CJ, Shaw PJ, Kirby J (2015) Gene expression signatures in motor neurone disease fibroblasts reveal dysregulation of metabolism, hypoxia-response and RNA processing functions. Neuropathol Appl Neurobiol 41:201–226. https://​doi.​org/​10.​1111/​nan.​12147 PubMedPubMedCentralCrossRef
159.
164.
Zurück zum Zitat Sasaki S, Iwata M (1996) Ultrastructural study of synapses in the anterior horn neurons of patients with amyotrophic lateral sclerosis. Neurosci Lett 204:53–56PubMedCrossRef Sasaki S, Iwata M (1996) Ultrastructural study of synapses in the anterior horn neurons of patients with amyotrophic lateral sclerosis. Neurosci Lett 204:53–56PubMedCrossRef
171.
Zurück zum Zitat Shannon KM, Bennett JP Jr, Friedman JH, The Pramipexole Study Group (1997) Efficacy of pramipexole, a novel dopamine agonist, as monotherapy in mild to moderate Parkinson’s disease. Neurology 49:724–728PubMedCrossRef Shannon KM, Bennett JP Jr, Friedman JH, The Pramipexole Study Group (1997) Efficacy of pramipexole, a novel dopamine agonist, as monotherapy in mild to moderate Parkinson’s disease. Neurology 49:724–728PubMedCrossRef
172.
Zurück zum Zitat Shaw PJ, Eggett CJ (2000) Molecular factors underlying selective vulnerability of motor neurons to neurodegeneration in amyotrophic lateral sclerosis. J Neurol 247(Suppl 1):I17–I27PubMedCrossRef Shaw PJ, Eggett CJ (2000) Molecular factors underlying selective vulnerability of motor neurons to neurodegeneration in amyotrophic lateral sclerosis. J Neurol 247(Suppl 1):I17–I27PubMedCrossRef
173.
Zurück zum Zitat Shefner JM, Cudkowicz ME, Schoenfeld D, Conrad T, Taft J, Chilton M, Urbinelli L, Qureshi M, Zhang H, Pestronk A, Caress J, Donofrio P, Sorenson E, Bradley W, Lomen-Hoerth C, Pioro E, Rezania K, Ross M, Pascuzzi R, Heiman-Patterson T, Tandan R, Mitsumoto H, Rothstein J, Smith-Palmer T, MacDonald D, Burke D, Consortium N (2004) A clinical trial of creatine in ALS. Neurology 63:1656–1661PubMedCrossRef Shefner JM, Cudkowicz ME, Schoenfeld D, Conrad T, Taft J, Chilton M, Urbinelli L, Qureshi M, Zhang H, Pestronk A, Caress J, Donofrio P, Sorenson E, Bradley W, Lomen-Hoerth C, Pioro E, Rezania K, Ross M, Pascuzzi R, Heiman-Patterson T, Tandan R, Mitsumoto H, Rothstein J, Smith-Palmer T, MacDonald D, Burke D, Consortium N (2004) A clinical trial of creatine in ALS. Neurology 63:1656–1661PubMedCrossRef
177.
Zurück zum Zitat Shimazu T, Hirschey MD, Newman J, He W, Shirakawa K, Le Moan N, Grueter CA, Lim H, Saunders LR, Stevens RD, Newgard CB, Farese RV Jr, de Cabo R, Ulrich S, Akassoglou K, Verdin E (2013) Suppression of oxidative stress by beta-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science 339:211–214. https://doi.org/10.1126/science.1227166 PubMedCrossRef Shimazu T, Hirschey MD, Newman J, He W, Shirakawa K, Le Moan N, Grueter CA, Lim H, Saunders LR, Stevens RD, Newgard CB, Farese RV Jr, de Cabo R, Ulrich S, Akassoglou K, Verdin E (2013) Suppression of oxidative stress by beta-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science 339:211–214. https://​doi.​org/​10.​1126/​science.​1227166 PubMedCrossRef
179.
Zurück zum Zitat Shults CW, Oakes D, Kieburtz K, Beal MF, Haas R, Plumb S, Juncos JL, Nutt J, Shoulson I, Carter J, Kompoliti K, Perlmutter JS, Reich S, Stern M, Watts RL, Kurlan R, Molho E, Harrison M, Lew M, Parkinson Study G (2002) Effects of coenzyme Q10 in early Parkinson disease: evidence of slowing of the functional decline. Arch Neurol 59:1541–1550PubMedCrossRef Shults CW, Oakes D, Kieburtz K, Beal MF, Haas R, Plumb S, Juncos JL, Nutt J, Shoulson I, Carter J, Kompoliti K, Perlmutter JS, Reich S, Stern M, Watts RL, Kurlan R, Molho E, Harrison M, Lew M, Parkinson Study G (2002) Effects of coenzyme Q10 in early Parkinson disease: evidence of slowing of the functional decline. Arch Neurol 59:1541–1550PubMedCrossRef
181.
Zurück zum Zitat Sills MA, Forsythe WI, Haidukewych D, MacDonald A, Robinson M (1986) The medium chain triglyceride diet and intractable epilepsy. Arch Dis Child 61:1168–1172PubMedPubMedCentralCrossRef Sills MA, Forsythe WI, Haidukewych D, MacDonald A, Robinson M (1986) The medium chain triglyceride diet and intractable epilepsy. Arch Dis Child 61:1168–1172PubMedPubMedCentralCrossRef
183.
Zurück zum Zitat So E, Mitchell JC, Memmi C, Chennell G, Vizcay-Barrena G, Allison L, Shaw CE, Vance C (2017) Mitochondrial abnormalities and disruption of the neuromuscular junction precede the clinical phenotype and motor neuron loss in hFUSWT transgenic mice. Hum Mol Genet. https://doi.org/10.1093/hmg/ddx415 So E, Mitchell JC, Memmi C, Chennell G, Vizcay-Barrena G, Allison L, Shaw CE, Vance C (2017) Mitochondrial abnormalities and disruption of the neuromuscular junction precede the clinical phenotype and motor neuron loss in hFUSWT transgenic mice. Hum Mol Genet. https://​doi.​org/​10.​1093/​hmg/​ddx415
186.
Zurück zum Zitat Stevens PR, Gawryluk JW, Hui L, Chen X, Geiger JD (2014) Creatine protects against mitochondrial dysfunction associated with HIV-1 Tat-induced neuronal injury. Curr HIV Res 12:378–387PubMedCrossRef Stevens PR, Gawryluk JW, Hui L, Chen X, Geiger JD (2014) Creatine protects against mitochondrial dysfunction associated with HIV-1 Tat-induced neuronal injury. Curr HIV Res 12:378–387PubMedCrossRef
189.
Zurück zum Zitat Tauskela JS (2007) MitoQ–a mitochondria-targeted antioxidant. IDrugs 10:399–412PubMed Tauskela JS (2007) MitoQ–a mitochondria-targeted antioxidant. IDrugs 10:399–412PubMed
192.
Zurück zum Zitat Thevenet J, De Marchi U, Domingo JS, Christinat N, Bultot L, Lefebvre G, Sakamoto K, Descombes P, Masoodi M, Wiederkehr A (2016) Medium-chain fatty acids inhibit mitochondrial metabolism in astrocytes promoting astrocyte-neuron lactate and ketone body shuttle systems. FASEB J 30:1913–1926. https://doi.org/10.1096/fj.201500182 PubMedCrossRef Thevenet J, De Marchi U, Domingo JS, Christinat N, Bultot L, Lefebvre G, Sakamoto K, Descombes P, Masoodi M, Wiederkehr A (2016) Medium-chain fatty acids inhibit mitochondrial metabolism in astrocytes promoting astrocyte-neuron lactate and ketone body shuttle systems. FASEB J 30:1913–1926. https://​doi.​org/​10.​1096/​fj.​201500182 PubMedCrossRef
200.
Zurück zum Zitat Vendelin M, Eimre M, Seppet E, Peet N, Andrienko T, Lemba M, Engelbrecht J, Seppet EK, Saks VA (2004) Intracellular diffusion of adenosine phosphates is locally restricted in cardiac muscle. Mol Cell Biochem 256–257:229–241PubMedCrossRef Vendelin M, Eimre M, Seppet E, Peet N, Andrienko T, Lemba M, Engelbrecht J, Seppet EK, Saks VA (2004) Intracellular diffusion of adenosine phosphates is locally restricted in cardiac muscle. Mol Cell Biochem 256–257:229–241PubMedCrossRef
201.
Zurück zum Zitat Vercruysse P, Sinniger J, El Oussini H, Scekic-Zahirovic J, Dieterle S, Dengler R, Meyer T, Zierz S, Kassubek J, Fischer W, Dreyhaupt J, Grehl T, Hermann A, Grosskreutz J, Witting A, Van Den Bosch L, Spreux-Varoquaux O, Ludolph AC, Dupuis L, Group GAS (2016) Alterations in the hypothalamic melanocortin pathway in amyotrophic lateral sclerosis. Brain 139:1106–1122. https://doi.org/10.1093/brain/aww004 PubMedCrossRef Vercruysse P, Sinniger J, El Oussini H, Scekic-Zahirovic J, Dieterle S, Dengler R, Meyer T, Zierz S, Kassubek J, Fischer W, Dreyhaupt J, Grehl T, Hermann A, Grosskreutz J, Witting A, Van Den Bosch L, Spreux-Varoquaux O, Ludolph AC, Dupuis L, Group GAS (2016) Alterations in the hypothalamic melanocortin pathway in amyotrophic lateral sclerosis. Brain 139:1106–1122. https://​doi.​org/​10.​1093/​brain/​aww004 PubMedCrossRef
203.
Zurück zum Zitat Vielhaber S, Winkler K, Kirches E, Kunz D, Buchner M, Feistner H, Elger CE, Ludolph AC, Riepe MW, Kunz WS (1999) Visualization of defective mitochondrial function in skeletal muscle fibers of patients with sporadic amyotrophic lateral sclerosis. J Neurol Sci 169:133–139PubMedCrossRef Vielhaber S, Winkler K, Kirches E, Kunz D, Buchner M, Feistner H, Elger CE, Ludolph AC, Riepe MW, Kunz WS (1999) Visualization of defective mitochondrial function in skeletal muscle fibers of patients with sporadic amyotrophic lateral sclerosis. J Neurol Sci 169:133–139PubMedCrossRef
207.
Zurück zum Zitat Wiedemann FR, Manfredi G, Mawrin C, Beal MF, Schon EA (2002) Mitochondrial DNA and respiratory chain function in spinal cords of ALS patients. J Neurochem 80:616–625PubMedCrossRef Wiedemann FR, Manfredi G, Mawrin C, Beal MF, Schon EA (2002) Mitochondrial DNA and respiratory chain function in spinal cords of ALS patients. J Neurochem 80:616–625PubMedCrossRef
208.
Zurück zum Zitat Wills AM, Hubbard J, Macklin EA, Glass J, Tandan R, Simpson EP, Brooks B, Gelinas D, Mitsumoto H, Mozaffar T, Hanes GP, Ladha SS, Heiman-Patterson T, Katz J, Lou JS, Mahoney K, Grasso D, Lawson R, Yu H, Cudkowicz M, Network MDACR (2014) Hypercaloric enteral nutrition in patients with amyotrophic lateral sclerosis: a randomised, double-blind, placebo-controlled phase 2 trial. Lancet 383:2065–2072. https://doi.org/10.1016/S0140-6736(14)60222-1 PubMedPubMedCentralCrossRef Wills AM, Hubbard J, Macklin EA, Glass J, Tandan R, Simpson EP, Brooks B, Gelinas D, Mitsumoto H, Mozaffar T, Hanes GP, Ladha SS, Heiman-Patterson T, Katz J, Lou JS, Mahoney K, Grasso D, Lawson R, Yu H, Cudkowicz M, Network MDACR (2014) Hypercaloric enteral nutrition in patients with amyotrophic lateral sclerosis: a randomised, double-blind, placebo-controlled phase 2 trial. Lancet 383:2065–2072. https://​doi.​org/​10.​1016/​S0140-6736(14)60222-1 PubMedPubMedCentralCrossRef
210.
Zurück zum Zitat Wong PC, Pardo CA, Borchelt DR, Lee MK, Copeland NG, Jenkins NA, Sisodia SS, Cleveland DW, Price DL (1995) An adverse property of a familial ALS-linked SOD1 mutation causes motor neuron disease characterized by vacuolar degeneration of mitochondria. Neuron 14:1105–1116PubMedCrossRef Wong PC, Pardo CA, Borchelt DR, Lee MK, Copeland NG, Jenkins NA, Sisodia SS, Cleveland DW, Price DL (1995) An adverse property of a familial ALS-linked SOD1 mutation causes motor neuron disease characterized by vacuolar degeneration of mitochondria. Neuron 14:1105–1116PubMedCrossRef
213.
Metadaten
Titel
Energy metabolism in ALS: an underappreciated opportunity?
verfasst von
Tijs Vandoorne
Katrien De Bock
Ludo Van Den Bosch
Publikationsdatum
16.03.2018
Verlag
Springer Berlin Heidelberg
Erschienen in
Acta Neuropathologica / Ausgabe 4/2018
Print ISSN: 0001-6322
Elektronische ISSN: 1432-0533
DOI
https://doi.org/10.1007/s00401-018-1835-x

Weitere Artikel der Ausgabe 4/2018

Acta Neuropathologica 4/2018 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.