Next Article in Journal
Efficacy and Immunological Impact of Intrathecal Baclofen Pumps in Managing Spasticity in Multiple Sclerosis in Adult Patients: A Systematic Review
Next Article in Special Issue
The Impact of α-Lipoic Acid Treatment on Multiple Sclerosis Disability: A Systematic Review and Meta-Analysis of Randomized Controlled Trials
Previous Article in Journal
Lifestyle, Cognition, and Disability Outcomes in Multiple Sclerosis: A Comprehensive Cohort Study
Previous Article in Special Issue
Resilience, Mental Health, Sleep, and Smoking Mediate Pathways Between Lifetime Stressors and Multiple Sclerosis Severity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

High-Fat Diet—Shared Environmental Risk Factor for Amyotrophic Lateral Sclerosis and Multiple Sclerosis

by
Thomas Gabriel Schreiner
1,2,*,
Liviu Iacob
2,
Cristina Georgiana Croitoru
2,
Diana Nicoleta Hodorog
1,2 and
Dan Iulian Cuciureanu
1,2
1
Department of Medical Specialties III, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
2
First Neurology Clinic, “N. Oblu” Clinical Emergency Hospital, 700309 Iasi, Romania
*
Author to whom correspondence should be addressed.
Sclerosis 2025, 3(1), 1; https://doi.org/10.3390/sclerosis3010001
Submission received: 8 December 2024 / Revised: 1 January 2025 / Accepted: 9 January 2025 / Published: 12 January 2025

Abstract

:
Background: Amyotrophic lateral sclerosis (ALS) and multiple sclerosis (MS) are, in essence, neurodegenerative disorders with significant individual, social, and economic burdens worldwide. Despite having different clinical onset and evolution, the two diseases share common risk factors and underlying pathophysiological mechanisms. Environmental risk factors are particularly interesting, considering the available effective counter strategies. High-fat diets remain a significant element that negatively impacts the onset and evolution of several disorders, including ALS and MS. Focusing on changeable disease-related aspects is increasingly appealing in the context of a lack of an effective treatment. Methods: This review aims to offer an updated overview of the influence of high-fat diets in modulating the risk of onset and progression of ALS and MS, based on the search of three relevant online databases. Results: In the first part, the shared pathophysiological mechanisms of ALS and MS are shown, and significant differences between the two disorders are highlighted. Subsequently, the most relevant research on this topic conducted in animal models and humans is presented, bringing additional proof of the critical role of high-fat diets in neurodegeneration. Finally, based on current knowledge, the authors offer potential therapeutic approaches and future relevant research directions to better control nutrition in ALS and MS patients, hoping to increase survival and quality of life. Conclusions: High-fat diets negatively impact the onset and evolution of ALS and MS.

1. Introduction

Neurodegenerative disorders (NDDs) are a heterogeneous group of diseases affecting the central nervous system (CNS) and are characterized by the progressive death of neurons that lead to the decrease and final loss of several motor, sensory, and cognitive functions [1]. While Alzheimer’s disease and Parkinson’s disease remain the most frequent NDDs [2], other pathologies such as amyotrophic lateral sclerosis (ALS) and, from a certain point of view regarding its evolution, multiple sclerosis (MS), are also of great interest considering their significant individual, social, and economic burden [3,4].
ALS, also known as Lou Gehrig’s disease, is the result of the progressive loss of upper and lower motor neurons, being clinically characterized by muscle weakness and eventually respiratory failure, with most patients being deceased within 2–3 years of symptom onset [5]. It affects approximately 2 per 100,000 people globally, with a lifetime risk of 1 in 350, with these figures expected to increase due to improved diagnostics and changing demographics [6]. Only 5–10% of the cases have a positive family history, with both genetic causes (such as SOD-1 [superoxide dismutase-1] and FUS [fused in sarcoma)] mutations) and environmental risk factors (smoking, pollution, heavy metals) [7] being considered for the majority of ALS patients. Besides the symptomatic treatment, only Riluzole, Edaravone, and Tofersen are approved for clinical use [8]; however, they have an unsatisfactory impact on quality of life and survival, with limited efficacy in modifying ALS history and preventing disease progression [9].
MS is traditionally known as the most common autoimmune demyelinating disorder of the CNS, affecting young adults (35.9 per 100,000 population) and leading to progressive neurodegeneration during its evolution [10]. Still, increasing evidence highlights its neurodegenerative aspects, based on the progressive neuronal loss, axonal degeneration, and brain atrophy observed in advanced MS stages, these being hallmarks shared with other NDDs. According to the most recent epidemiological data, the global incidence is 2.1 per 100,000 persons/year, and the worldwide prevalence is over 2.8 million MS patients, with figures expected to rise in the near future [11]. This disorder also imposes high socioeconomic costs on healthcare systems [12]. For example, in 2022 in the United States, the MS-related total economic burden was USD 85.4 billion, with a direct medical cost of USD 63.3 billion [13]. MS significantly impacts patients’ quality of life, causing diverse neurological symptoms: vision and balance problems, motor and sensory deficits, urinary problems, and cognitive impairment, the heterogeneous clinical picture being explained by the demyelinating plaques that can affect various CNS regions. In the last decades, there has been an effervescence of disease-modifying therapies (DMTs) that have shown efficacy in counteracting the clinical and radiological activity of MS; still, DMTs have little to no effect on disease progression independent of such acute episodes and on the underlying neurodegenerative processes [14].
While ALS and MS are different entities in clinical presentation, underlying pathophysiology, and treatment opportunities, there are still shared risk factors and molecular mechanisms that have raised researchers’ interest in recent years. One relevant example is the impact of high-fat diet in the onset and evolution of both disorders in the context of the increasing importance of nutrition and nutritional interventions in modulating neurological disorders, most probably via the intensely discussed gut–brain axis [15,16]. In this context, this review aims to offer an updated overview of the role of high-fat diet in ALS and MS risk and progression. The shared pathophysiological mechanisms of ALS and MS are presented in the first part, together with a comparative analysis of the significant differences between the two pathologies. Subsequently, the authors highlight the most relevant research on this topic conducted in animal models and humans, bringing additional proof of the negative impact of a high-fat diet on neurodegeneration. In the final part, based on the currently available knowledge, potential therapeutic approaches and future relevant research directions used to better control nutrition in ALS and MS patients are offered, with the hope of increasing the survival and the quality of life of these patients.

2. Materials and Method

Despite this not being a systematic review, to include in this work the most relevant studies related to high-fat diets’ impact on ALS and MS, the authors conducted a systematic search in the most relevant online databases, namely, PubMed/Medline (https://pubmed.ncbi.nlm.nih.gov/, accessed on 30 November 2024), ScienceDirect (https://www.sciencedirect.com/, accessed on 30 November 2024), and Google Scholar (https://scholar.google.com/, accessed on 30 November 2024). To cover all significant aspects (pathophysiological mechanisms, trials in animal models, clinical trials) covered in the review, different combinations of the following relevant terms were used: ‘amyotrophic lateral sclerosis’, ‘multiple sclerosis’, ‘high-fat diet’, ‘neuroinflammation’, ‘oxidative stress’, ‘neurodegenerative disease’. The inclusion criteria comprised original, peer-reviewed English language studies conducted on patients and in silico, in vitro, and in vivo models, published up to the present. In addition, some narrative and systematic reviews were also included, mainly when covering aspects lacking original research. The authors admit that this search and inclusion strategy might lead to the exclusion of some potentially valuable studies, which represents the main limitation of this paper.

3. Comparative Overview of ALS and MS Pathophysiological Mechanisms

According to the classical definition, ALS is a progressive NDD characterized by the dysfunction of both upper and lower motor neurons, being long considered a motor system disease. Despite its typical focal beginning in the upper or lower limbs and bulbar or respiratory regions, ALS progresses causing widespread muscle weakness, with a restrictive respiratory failure marking its terminal stage [17]. Understanding the link between upper and lower motor neuron dysfunction is crucial for deciphering ALS pathogenesis, with three main theories currently existing: the dying-forward hypothesis (that suggests that ALS originates in the cortex and spreads to lower neurons), the dying-back hypothesis (that proposes the beginning of the degeneration in the lower motor neuron), and the independent hypothesis (which states that upper and lower motor neuron degenerate independently in a random and contiguous pattern) [18].
While its etiology is still incompletely elucidated, the onset and evolution of ALS appear to result from a complex interaction between genetic and environmental factors, which subsequently leads to disruptions in critical molecular pathways and sustained neurodegeneration. Research suggests a multi-step phenomenon with up to six consecutive phases/factors necessary for the development of ALS-related neurodegeneration [19]. Among the genetic contributors, mutations in the C9orf72, TARDBP (encoding TDP-43), and fused in sarcoma (FUS) genes lead to the dysregulation of RNA metabolism, resulting in abnormal protein translation and the formation of intracellular neuronal aggregates [20]. Mutations in the SOD-1 gene further exacerbate neurodegenerative processes by inducing oxidative stress, causing mitochondrial dysfunction and leading to defective axonal transport with the formation of subsequent intracellular aggregates [21]. These genetic disturbances intersect with critical pathogenic pathways for ALS, such as glutamate excitotoxicity [22]. The dysfunction of the astrocytic excitatory amino acid transporter 2 (EAAT2) leads to reduced glutamate uptake from the synaptic cleft, and excess glutamate subsequently activates Ca2+-dependent enzymatic pathways [23]. Environmental factors, including toxins and metabolic stressors, may also exacerbate these molecular disruptions, promoting neuroinflammation. Activated microglia release proinflammatory cytokines that amplify neurotoxicity and contribute to disease progression [24].
Regarding environmental factors, diet remains a highly discussed issue, with micro- and macronutrients from different diets being hypothetically linked to many pathological modifications in ALS. Several environmental toxins, such as β-methylamino-L-alanine (BMAA) found in seafood and cycad seeds, have been linked to ALS, particularly in regions like Guam and the Kii peninsula of Japan [25]. Additionally, excessive consumption of high-fat foods, particularly in countries like Sweden and Scotland, is associated with a higher incidence of ALS, although the exact role of dietary fat remains debated [26]. Meanwhile, oxidative stress and mitochondrial dysfunction are known to play critical roles in the disease’s progression, with evidence suggesting that diets rich in antioxidants may help reduce oxidation. Curcumin, creatine, and coenzyme Q10 (CoQ10) have shown promise in animal studies for their antioxidant properties, but their efficacy in humans remains uncertain [27]. Dietary sources of heavy metals such as lead, cadmium, and mercury have also been studied for their potential connection to ALS [28,29]. Bioaccumulation and biomagnification in the food chain, particularly in seafood, could lead to increased exposure to these metals, which are suspected to promote neurodegeneration through oxidative damage and excitotoxicity. Although evidence linking mercury exposure to ALS is inconclusive, some studies suggest that individuals with ALS might be more susceptible to toxic metals due to genetic predispositions [30]. The overall role of diet in ALS is complex, with factors such as antioxidant intake potentially slowing disease progression, while exposure to neurotoxins could increase the risk of disease onset.
Contrary to ALS, the pathophysiology of MS seems to be better understood, although incompletely elucidated. This autoimmune disease is characterized by the formation of plaques in the CNS, leading to neuroinflammation, demyelination, axonal damage, and neuronal loss. The disease results from autoreactive immune cells, particularly CD8+ T cells, CD4+ Th1 cells, and Th17 cells, crossing the blood–brain barrier (BBB) and triggering autoimmune reactions in the CNS [31]. The failure of peripheral immune tolerance, possibly due to impaired regulatory T cells and dysfunctional autoreactive T cells, contributes to MS development. Additionally, B cells play a crucial role by producing oligoclonal immunoglobulins (OCBs) in the cerebrospinal fluid (CSF), with memory B cells significantly contributing to disease progression [32]. According to the viral hypothesis, infected B cells may also serve as ‘reservoirs’ for latent viral infections, such as by Epstein–Barr Virus (EBV), which was demonstrated to play a significant role in MS pathogenesis [33]. EBV-infected B cells not only evade immune control systems and apoptosis but also become capable of more efficiently presenting antigens to CD8+ T cells. Microglia and macrophages contribute to neurodegeneration by releasing cytokines like tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β), promoting oxidative stress, excitotoxicity, and mitochondrial damage [34]. However, microglia were also shown to have anti-inflammatory properties, aiding in remyelination [35]. While MS onset results from the mixed impact of genetic and environmental factors, disease progression results from the complex interactions between immune cells, latent viral infections, and inflammatory processes.
Similar to ALS, studies on dietary interventions in MS are still to reach a clear conclusion. Although no specific diet is scientifically proven to treat MS, certain foods and supplements are recommended for their potential benefits. These include fish, low-fat foods, whole grains, vitamins, and omega-3 fatty acids [36]. Some evidence suggests that carotenoids, polyphenols, and other bioactive molecules from vegetables may have anti-inflammatory and antioxidant effects [37]. Vitamin D deficiency is recognized as a risk factor for MS [38], while the role of low levels of vitamin B12, initially thought to be linked to increased disability in MS patients, still remains elusive [39]. Although diets and alternative therapies cannot replace conventional MS treatments, a healthy diet might reduce inflammation and improve physical deficits, potentially serving as a complementary therapy. Further research is required to determine how dietary supplementation can slow MS progression and delay its onset.
ALS and MS share common pathophysiological pathways, including oxidative stress and neuroinflammation, which contribute to neuronal damage in both conditions. Oxidative stress, characterized by an imbalance between reactive oxygen species and antioxidant defenses, leads to cellular injury and mitochondrial dysfunction, critical neurodegeneration components in both MS and ALS [40,41]. In MS, oxidative stress is closely tied to the inflammatory environment generated by pathologically reactive immune cells, resulting in demyelination, axonal injury, and the formation of plaques [42]. Similarly, in ALS, oxidative damage is associated with motor neuron death, although the sources of oxidative stress may differ, with protein misfolding, mitochondrial dysfunction, and excitotoxicity playing more significant roles [43]. Neuroinflammation is a shared feature of both diseases, with microglial activation contributing to disease progression [44]. In MS, activated microglia release pro-inflammatory cytokines and reactive species, exacerbating the immune-mediated destruction of myelin [45], while in ALS, microglial activation leads to neurotoxic effects on motor neurons, perpetuating their degeneration [46].
Despite these shared mechanisms, highlighted in Figure 1, the two diseases diverge significantly in their specific pathophysiological processes and damaged neural structures. MS is primarily an autoimmune disorder orchestrated by T and B cells [47]. The pathological hallmark of MS is the formation of demyelinating plaques, where inflammatory cells infiltrate and cause destruction, impairing the transmission of nerve signals [48]. In contrast, ALS is predominantly a motor neuron disease characterized by the progressive degeneration of upper and low motor neurons. In ALS, neurodegeneration is driven by mechanisms such as glutamate excitotoxicity, mitochondrial dysfunction, and the accumulation of toxic proteins like SOD-1, contributing to motor neuron death [49].
Understanding the similarities and differences between the pathophysiological processes of MS and ALS can be intuitive for comprehending how high-fat diets might negatively impact and exacerbate these disorders. In the following sections, the authors reveal the most relevant studies that prove the role of high-fat diets in the onset and development of MS- and ALS-related neurodegeneration, subsequently showing how dietary changes might help slow disease progression.

4. Relevant Research in Animal Models and Humans

4.1. High-Fat Diets and MS

Research on the association between high-fat diets and MS has been conducted in both animal models and humans. Table 1 summarizes the most relevant research on this topic, highlighting the studies’ methodology, limitations, and conclusions.
Despite its limitations, the experimental autoimmune encephalomyelitis (EAE) mouse model is a frequently used animal model for MS-related research, playing a role also in studies on the high-fat diet–MS association. EAE mice receiving a high-fat diet are precious models for a more in-depth understanding of the pathophysiological mechanisms underlying this association. Research conducted by Timmermans et al. showed the significant role of the activated renin–angiotensin system, the increased pro-inflammatory response, and the impact of infiltrated immune cells resulting from the high intake of dietary fats [52]. More recently, fundamental studies demonstrated the increasing role of the gut–brain axis in the correlation between fat diet and MS, the alteration of intestinal microbiota and intestinal permeability modulating the systemic inflammation that subsequently sustains the CNS inflammatory state [51]. Another relevant structure, the blood–brain barrier, also suffers structural and functional damage. This leads to high levels of inflammatory cells entering the CNS, favoring the formation and aggravation of the demyelinating plaques [50].
The preliminary conclusions from the studies conducted in animal models are still to be confirmed in humans via larger cohorts of MS patients. Despite the comprehensive literature search, the authors found only one relevant research study that specifically addressed the role of high-fat diets as a risk factor for MS onset and progression. In this regard, the study conducted by Asgharzadeh et al. is worth mentioning, with the group of MS patients registering a significantly higher high-fat food consumption compared to the healthy controls [53]. As this observation is only correlational, there is an increasing need for further, more extensive cohort studies and interventional studies to confirm the correlation between MS and high-fat food intake.

4.2. High-Fat Diets and ALS

The issue of high-fat diets in ALS is more complex than in MS, with the negative high-fat diet–MS association. Given the catabolic nature of the changes in the metabolism of ALS patients, a high-fat diet seems to play a dual role: in the first stages of the disease, high-fat diets may sustain neuroinflammation and increase oxidative stress, playing a pivotal role as a risk factor for the development and advancement of the disease, while in the final stages of ALS, a high-fat diet might be beneficial, restoring biological imbalances and even slightly extending the lifespan. The metabolic dysfunctions found in the most frequently used ALS mouse models comprise reduced body weight, reduced fat and muscle mass, and an imbalanced caloric intake. Thus, a high-caloric intake, including a high-fat diet, could be an option for improving the general condition of these animals. In this regard, worth mentioning is the research conducted by Coughlan et al., in which a novel ALS mouse model received a high-fat jelly diet, with a promising positive impact on sudden death prevention and survival extension [54]. Several studies were conducted on the implications of hypercaloric diets in ALS mouse models, with results showing the potential for improvement of the nutritional status and prolonged life [55].
Regarding research conducted on ALS patients, the findings on the role of lipids in ALS prognosis are conflicting. On the one hand, higher frequencies of hyperlipidemia, increased triglycerides, and altered cholesterol ratios were reported in ALS patients compared to controls, with higher cholesterol levels associated with more prolonged survival [56]. On the other hand, no clear association between lipid levels and ALS survival has been established, while some authors linked dyslipidemia to worsened respiratory function [56]. Worth mentioning is the work of Dorst et al. (see Table 2), where the authors compared a high-fat diet to other high-caloric dietary supplementations, assessing the patient’s tolerability and the impact on weight gain [57]. Several similar studies were conducted in the last decade, with mixed results [58]; however, the focus was on hypercaloric intake, with no differentiation of high-fat diets, making it challenging to draw relevant conclusions.

5. Dietary Changes as Potential Therapeutic Interventions

Despite the incomplete understanding of the physiology of MS and ALS and the conflicting results regarding the dual role of high-fat diets in ALS, dietary changes have long been regarded as adjuvant therapeutic measures with increased potential (see Table 3). The proof is the many trials conducted in recent years, particularly in MS patients. For example, the work of Villa et al. highlighted the benefits of low-saturated-fat (Swank) and modified Paleolithic elimination (Wahls) diets on weight, cholesterol levels, and perceived fatigue in MS patients [59]. More interesting are the results obtained by Dean et al. in their pilot study, despite the small number (n = 12) of initially included patients and the high dropout rate (50%), where patients followed a low-saturated-fat diet for the median duration of 37 months. The patients who were able to follow the diet registered a decrease in their EDSS scores, no relapses, and a stable disease according to the imaging criteria of yearly magnetic resonance imaging (MRI) scans [60]. According to a systematic review, plant-based diets were demonstrated to be helpful in MS patients, together with low-fat and ketogenic diets [61]. Moreover, a Cochrane review from 2020 addressing the influences of dietary interventions on MS-related outcomes demonstrated the most relevant directions of the dietary interventions used in MS patients [62]. According to recent works, diet might also improve complementary symptoms in MS patients, such as fatigue, improving the patient’s quality of life [63,64]. Most clinical trials focused on supplementation to increase polyunsaturated fatty acids (PUFAs), the administration of antioxidant supplements, or dietary programs. Other nutritional supplements such as biotin, acetyl L-carnitine, riboflavin, probiotics, and palmitoylethanolamide were also tested in smaller trials. There is uncertain evidence on whether PUFA administration modulates the relapse rate, disability, or overall clinical status in MS patients. Similarly, insufficient evidence exists to determine whether supplementation with antioxidants or other dietary interventions significantly impacts MS.
When considering ALS, certain foods and compounds might prevent ALS onset or slightly slow its progression, mainly by countering the significant factors of neuronal degeneration, oxidative stress, and chronic inflammation. Several nutraceuticals share common pathophysiological pathways with different types of fats, becoming potential therapeutic strategies that may mitigate the adverse effects of a high-fat diet. Among the promising nutraceuticals intensely studied in recent years, curcumin showed promising effects due to its antioxidant and anti-inflammatory properties. While animal studies suggest that increasing the levels of antioxidants like glutathione and curcumin potentially reduces neuroinflammation and oxidative stress, interventions in humans have led to mixed results [65]. Some trials have shown that curcumin, in combination with Riluzole, may extend survival in ALS patients, though side effects like nausea have been noted [66]. Moreover, dimethoxy curcumin has shown protective effects on mitochondrial function in cellular ALS models, explaining mitochondria’s role in ALS development and a possible action pathway for curcumin [67]. Other supplements currently under investigation in ALS patients are creatine and CoQ10. Creatine has been shown to offer neuroprotection in animal models, but human studies report only limited improvements, with further research needed [68]. CoQ10, an essential antioxidant to mitochondrial function, was tested in various neurodegenerative conditions, including ALS [69]. Though generally safe, CoQ10 may interact with certain drugs and cause gastrointestinal side effects [70]. Other antioxidants, such as vitamins C and E, have mixed evidence for neuroprotection in ALS. Ongoing research also involves the role of phytochemicals such as carotenoids, polyphenols, and terpenoids as antioxidant compounds that could help reduce neuroinflammation and prevent neuronal damage [71]. Resveratrol, a polyphenol, has demonstrated cognitive benefits by increasing sirtuin 1 (SIRT1) levels, a gene regulator, while terpenoids showed antioxidative potential in vitro [72]. Despite promising findings, establishing the effectiveness of these phytochemicals in ALS requires more in-depth research on larger cohorts of patients.

6. Conclusions and Future Research Directions

ALS and MS remain two relevant pathologies in the heterogeneous group of NDDs, with their prevalence expected to increase in the following decades. Despite intensive research regarding the causative factor leading to neurodegeneration, there are still many unknowns, and an effective disease-stopping therapy is lacking. Still, several risk factors are known, with environmental factors such as high-fat diet having appeared as a prominent research topic in recent years. In MS, high-fat diets have without doubt a negative impact on the disease onset and evolution; however, the relation between high-fat diet and ALS is dependent on ALS stage: in earlier phases, high-fat diets increase oxidative stress and neuroinflammation, sustaining the rapid neurodegeneration of the motor neurons, while in the terminal stages, high-fat diets, together with other high-caloric diets, seem to prolong disease duration.
Based on the current literature on the impact of a high-fat diet in ALS and MS, there are at least three directions that future studies should focus on in the near future. Regarding studies in humans, large-cohort, randomized clinical trials are mandatorily needed to assess the real impact of high-fat diets on the onset and evolution of MS and, particularly, in the later stages of ALS. Also, as high-fat diets differ in composition (types of fat, absolute percentages), future studies in humans should deliver clearer methodologies to follow the impact of saturated/unsaturated/trans fats on the pathophysiological processes. Simultaneously, fundamental research in cellular and animal models should be continued, with the scope of a more in-depth understanding of the molecular pathways involved. Finally, the most promising area of research is related to the dietary changes and multiple diets that should thoroughly be studied in MS and ALS patients as add-on therapies that may significantly impact patients’ quality of life and disease duration.

Author Contributions

Conceptualization, T.G.S., L.I. and D.I.C.; methodology, data collection, and formal analysis, T.G.S., C.G.C. and D.N.H.; writing—original draft preparation, T.G.S., L.I. and D.N.H.; writing—review and editing, C.G.C. and D.I.C.; supervision, D.I.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Lamptey, R.N.L.; Chaulagain, B.; Trivedi, R.; Gothwal, A.; Layek, B.; Singh, J. A Review of the Common Neurodegenerative Disorders: Current Therapeutic Approaches and the Potential Role of Nanotherapeutics. Int. J. Mol. Sci. 2022, 23, 1851. [Google Scholar] [CrossRef] [PubMed]
  2. Tanaka, M.; Toldi, J.; Vécsei, L. Exploring the Etiological Links behind Neurodegenerative Diseases: Inflammatory Cytokines and Bioactive Kynurenines. Int. J. Mol. Sci. 2020, 21, 2431. [Google Scholar] [CrossRef] [PubMed]
  3. Boentert, M.; Hermann, A.; Großkreutz, J. Amyotrophic Lateral Sclerosis: Advances and Prospects. J. Clin. Med. 2023, 12, 5055. [Google Scholar] [CrossRef] [PubMed]
  4. Sarhan, A.A.; El-Sharkawy, K.A.; Mahmoudy, A.M.; Hashim, N.A. Burden of multiple sclerosis: Impact on the patient, family and society. Mult. Scler. Relat. Disord. 2022, 63, 103864. [Google Scholar] [CrossRef]
  5. Mead, R.J.; Shan, N.; Reiser, H.J.; Marshall, F.; Shaw, P.J. Amyotrophic lateral sclerosis: A neurodegenerative disorder poised for successful therapeutic translation. Nat. Rev. Drug Discov. 2023, 22, 185–212. [Google Scholar] [CrossRef] [PubMed]
  6. Longinetti, E.; Fang, F. Epidemiology of amyotrophic lateral sclerosis: An update of recent literature. Curr. Opin. Neurol. 2019, 32, 771–776. [Google Scholar] [CrossRef]
  7. Duan, Q.Q.; Jiang, Z.; Su, W.M.; Gu, X.J.; Wang, H.; Cheng, Y.F. Risk factors of amyotrophic lateral sclerosis: A global meta-summary. Front. Neurosci. 2023, 17, 1177431. [Google Scholar] [CrossRef] [PubMed]
  8. Blair, H.A. Tofersen: First Approval. Drugs 2023, 83, 1039–1043. [Google Scholar] [CrossRef] [PubMed]
  9. Hoxhaj, P.; Hastings, N.; Kachhadia, M.P.; Gupta, R.; Sindhu, U.; Durve, S.A.; Azam, A.; Vinueza, M.J.; Win, S.H.; Rathod, D.C.; et al. Exploring Advancements in the Treatment of Amyotrophic Lateral Sclerosis: A Comprehensive Review of Current Modalities and Future Prospects. Cureus 2023, 15, e45489. [Google Scholar] [CrossRef] [PubMed]
  10. Wallin, M.T.; Culpepper, W.J.; Campbell, J.D.; Nelson, L.M.; Langer-Gould, A.; Marrie, R.A.; Cutter, G.R.; Kaye, W.E.; Wagner, L.; Tremlett, H.; et al. The prevalence of MS in the United States: A population-based estimate using health claims data. Neurology 2019, 92, e1029–e1040, Erratum in Neurology 2019, 93, 688. [Google Scholar] [CrossRef]
  11. Walton, C.; King, R.; Rechtman, L.; Kaye, W.; Leray, E.; Marrie, R.A.; Robertson, N.; La Rocca, N.; Uitdehaag, B.; van Der Mei, I.; et al. Rising prevalence of multiple sclerosis worldwide: Insights from the Atlas of MS, third edition. Mult. Scler. 2020, 26, 1816–1821. [Google Scholar] [CrossRef] [PubMed]
  12. Dillon, P.; Heer, Y.; Karamasioti, E.; Rouzic, E.M.-L.; Marcelli, G.; Di Maio, D.; Braune, S.; Kobelt, G.; Wasem, J. The socioeconomic impact of disability progression in multiple sclerosis: A retrospective cohort study of the German NeuroTransData (NTD) registry. Mult. Scler. J. Exp. Transl. Clin. 2023, 9, 20552173231187810. [Google Scholar] [CrossRef] [PubMed]
  13. Bebo, B.; Cintina, I.; LaRocca, N.; Ritter, L.; Talente, B.; Hartung, D.; Ngorsuraches, S.; Wallin, M.; Yang, G. The Economic Burden of Multiple Sclerosis in the United States: Estimate of Direct and Indirect Costs. Neurology 2022, 98, e1810–e1817. [Google Scholar] [CrossRef] [PubMed]
  14. Hauser, S.L.; Cree, B.A.C. Treatment of Multiple Sclerosis: A Review. Am. J. Med. 2020, 133, 1380–1390.e2. [Google Scholar] [CrossRef]
  15. Allogmanny, S.; Probst, Y. Dietary Modification Combined with Nutrition Education and Counseling for Metabolic Comorbidities in Multiple Sclerosis: Implications for Clinical Practice and Research. Curr. Nutr. Rep. 2024, 13, 106–112. [Google Scholar] [CrossRef] [PubMed]
  16. D’Antona, S.; Caramenti, M.; Porro, D.; Castiglioni, I.; Cava, C. Amyotrophic Lateral Sclerosis: A Diet Review. Foods 2021, 10, 3128. [Google Scholar] [CrossRef] [PubMed]
  17. Niedermeyer, S.; Murn, M.; Choi, P.J. Respiratory Failure in Amyotrophic Lateral Sclerosis. Chest 2019, 155, 401–408. [Google Scholar] [CrossRef]
  18. Eisen, A.; Vucic, S.; Mitsumoto, H. History of ALS and the competing theories on pathogenesis: IFCN handbook chapter. Clin. Neurophysiol. Pract. 2023, 9, 1–12. [Google Scholar] [CrossRef] [PubMed]
  19. Chiò, A.; Mazzini, L.; D’Alfonso, S.; Corrado, L.; Canosa, A.; Moglia, C.; Manera, U.; Bersano, E.; Brunetti, M.; Barberis, M.; et al. The multistep hypothesis of ALS revisited: The role of genetic mutations. Neurology 2018, 91, e635–e642. [Google Scholar] [CrossRef]
  20. Wang, H.; Guan, L.; Deng, M. Recent progress of the genetics of amyotrophic lateral sclerosis and challenges of gene therapy. Front. Neurosci. 2023, 17, 1170996. [Google Scholar] [CrossRef]
  21. Berdyński, M.; Miszta, P.; Safranow, K.; Andersen, P.M.; Morita, M.; Filipek, S.; Żekanowski, C.; Kuźma-Kozakiewicz, M. SOD1 mutations associated with amyotrophic lateral sclerosis analysis of variant severity. Sci. Rep. 2022, 12, 103. [Google Scholar] [CrossRef] [PubMed]
  22. Arnold, F.J.; Putka, A.F.; Raychaudhuri, U.; Hsu, S.; Bedlack, R.S.; Bennett, C.L.; La Spada, A.R. Revisiting Glutamate Excitotoxicity in Amyotrophic Lateral Sclerosis and Age-Related Neurodegeneration. Int. J. Mol. Sci. 2024, 25, 5587. [Google Scholar] [CrossRef] [PubMed]
  23. Rosenblum, L.T.; Trotti, D. EAAT2 and the Molecular Signature of Amyotrophic Lateral Sclerosis. Adv. Neurobiol. 2017, 16, 117–136. [Google Scholar] [CrossRef] [PubMed]
  24. You, J.; Youssef, M.M.M.; Santos, J.R.; Lee, J.; Park, J. Microglia and Astrocytes in Amyotrophic Lateral Sclerosis: Disease-Associated States, Pathological Roles, and Therapeutic Potential. Biology 2023, 12, 1307. [Google Scholar] [CrossRef]
  25. Bradley, W.G.; Mash, D.C. Beyond Guam: The cyanobacteria/BMAA hypothesis of the cause of ALS and other neurodegenerative diseases. Amyotroph. Lateral Scler. 2009, 10 (Suppl. S2), 7–20. [Google Scholar] [CrossRef] [PubMed]
  26. D’Amico, E.; Grosso, G.; Nieves, J.W.; Zanghì, A.; Factor-Litvak, P.; Mitsumoto, H. Metabolic Abnormalities, Dietary Risk Factors and Nutritional Management in Amyotrophic Lateral Sclerosis. Nutrients 2021, 13, 2273. [Google Scholar] [CrossRef] [PubMed]
  27. Daverey, A.; Agrawal, S.K. Neuroprotective effects of Riluzole and Curcumin in human astrocytes and spinal cord white matter hypoxia. Neurosci. Lett. 2020, 738, 135351. [Google Scholar] [CrossRef]
  28. Peters, S.; Broberg, K.; Gallo, V.; Levi, M.; Kippler, M.; Vineis, P.; Veldink, J.; van Den Berg, L.; Middleton, L.; Travis, R.C.; et al. Blood Metal Levels and Amyotrophic Lateral Sclerosis Risk: A Prospective Cohort. Ann. Neurol. 2021, 89, 125–133. [Google Scholar] [CrossRef]
  29. Oggiano, R.; Pisano, A.; Sabalic, A.; Farace, C.; Fenu, G.; Lintas, S.; Forte, G.; Bocca, B.; Madeddu, R. An overview on amyotrophic lateral sclerosis and cadmium. Neurol. Sci. 2021, 42, 531–537. [Google Scholar] [CrossRef]
  30. Figueroa-Romero, C.; Mikhail, K.A.; Gennings, C.; Curtin, P.; Bello, G.A.; Botero, T.M.; Goutman, S.A.; Feldman, E.L.; Arora, M.; Austin, C. Early life metal dysregulation in amyotrophic lateral sclerosis. Ann. Clin. Transl. Neurol. 2020, 7, 872–882. [Google Scholar] [CrossRef]
  31. Moser, T.; Akgün, K.; Proschmann, U.; Sellner, J.; Ziemssen, T. The role of TH17 cells in multiple sclerosis: Therapeutic implications. Autoimmun. Rev. 2020, 19, 102647. [Google Scholar] [CrossRef]
  32. Comi, G.; Bar-Or, A.; Lassmann, H.; Uccelli, A.; Hartung, H.P.; Montalban, X.; Sørensen, P.S.; Hohlfeld, R.; Hauser, S.L. Role of B Cells in Multiple Sclerosis and Related Disorders. Ann. Neurol. 2021, 89, 13–23. [Google Scholar] [CrossRef]
  33. Soldan, S.S.; Lieberman, P.M. Epstein-Barr virus and multiple sclerosis. Nat. Rev. Microbiol. 2023, 21, 51–64. [Google Scholar] [CrossRef]
  34. Zhang, X.; Chen, F.; Sun, M.; Wu, N.; Liu, B.; Yi, X.; Ge, R.; Fan, X. Microglia in the context of multiple sclerosis. Front. Neurol. 2023, 14, 1157287. [Google Scholar] [CrossRef]
  35. Guerrero, B.L.; Sicotte, N.L. Microglia in Multiple Sclerosis: Friend or Foe? Front. Immunol. 2020, 11, 374. [Google Scholar] [CrossRef] [PubMed]
  36. Tryfonos, C.; Mantzorou, M.; Fotiou, D.; Vrizas, M.; Vadikolias, K.; Pavlidou, E.; Giaginis, C. Dietary Supplements on Controlling Multiple Sclerosis Symptoms and Relapses: Current Clinical Evidence and Future Perspectives. Medicines 2019, 6, 95. [Google Scholar] [CrossRef]
  37. Kabir, M.T.; Rahman, M.H.; Shah, M.; Jamiruddin, M.R.; Basak, D.; Al-Harrasi, A.; Bhatia, S.; Ashraf, G.M.; Najda, A.; El-Kott, A.F.; et al. Therapeutic promise of carotenoids as antioxidants and anti-inflammatory agents in neurodegenerative disorders. Biomed. Pharmacother. 2022, 146, 112610. [Google Scholar] [CrossRef] [PubMed]
  38. Gombash, S.E.; Lee, P.W.; Sawdai, E.; Lovett-Racke, A.E. Vitamin D as a Risk Factor for Multiple Sclerosis: Immunoregulatory or Neuroprotective? Front. Neurol. 2022, 13, 796933. [Google Scholar] [CrossRef] [PubMed]
  39. Najafi, M.R.; Shaygannajad, V.; Mirpourian, M.; Gholamrezaei, A. Vitamin B(12) Deficiency and Multiple Sclerosis; Is there Any Association? Int. J. Prev. Med. 2012, 3, 286–289. [Google Scholar] [PubMed]
  40. Park, H.R.; Yang, E.J. Oxidative Stress as a Therapeutic Target in Amyotrophic Lateral Sclerosis: Opportunities and Limitations. Diagnostics 2021, 11, 1546. [Google Scholar] [CrossRef] [PubMed]
  41. Jiménez-Jiménez, F.J.; Alonso-Navarro, H.; Salgado-Cámara, P.; García-Martín, E.; Agúndez, J.A.G. Oxidative Stress Markers in Multiple Sclerosis. Int. J. Mol. Sci. 2024, 25, 6289. [Google Scholar] [CrossRef] [PubMed]
  42. Sanabria-Castro, A.; Alape-Girón, A.; Flores-Díaz, M.; Echeverri-McCandless, A.; Parajeles-Vindas, A. Oxidative stress involvement in the molecular pathogenesis and progression of multiple sclerosis: A literature review. Rev. Neurosci. 2024, 35, 355–371. [Google Scholar] [CrossRef] [PubMed]
  43. Motataianu, A.; Serban, G.; Barcutean, L.; Balasa, R. Oxidative Stress in Amyotrophic Lateral Sclerosis: Synergy of Genetic and Environmental Factors. Int. J. Mol. Sci. 2022, 23, 9339. [Google Scholar] [CrossRef] [PubMed]
  44. Li, G.; Esiri, M.M.; Ansorge, O.; DeLuca, G.C. Concurrent multiple sclerosis and amyotrophic lateral sclerosis: Where inflammation and neurodegeneration meet? J. Neuroinflamm. 2012, 9, 20. [Google Scholar] [CrossRef] [PubMed]
  45. Mado, H.; Adamczyk-Sowa, M.; Sowa, P. Role of Microglial Cells in the Pathophysiology of MS: Synergistic or Antagonistic? Int. J. Mol. Sci. 2023, 24, 1861. [Google Scholar] [CrossRef] [PubMed]
  46. Clarke, B.E.; Patani, R. The microglial component of amyotrophic lateral sclerosis. Brain 2020, 143, 3526–3539. [Google Scholar] [CrossRef] [PubMed]
  47. van Langelaar, J.; Rijvers, L.; Smolders, J.; van Luijn, M.M. B and T Cells Driving Multiple Sclerosis: Identity, Mechanisms and Potential Triggers. Front. Immunol. 2020, 11, 760. [Google Scholar] [CrossRef] [PubMed]
  48. Coutinho Costa, V.G.; Araújo, S.E.; Alves-Leon, S.V.; Gomes, F.C.A. Central nervous system demyelinating diseases: Glial cells at the hub of pathology. Front. Immunol. 2023, 14, 1135540. [Google Scholar] [CrossRef]
  49. Hu, Y.; Chen, W.; Wei, C.; Jiang, S.; Li, S.; Wang, X.; Xu, R. Pathological mechanisms of amyotrophic lateral Sclerosis. Neural Regen. Res. 2024, 19, 1036–1044. [Google Scholar] [CrossRef]
  50. Davanzo, G.G.; Castro, G.; Monteiro, L.B.; Castelucci, B.G.; Jaccomo, V.H.; da Silva, F.C.; Marques, A.M.; Francelin, C.; de Campos, B.B.; de Aguiar, C.F.; et al. Obesity increases blood-brain barrier permeability and aggravates the mouse model of multiple sclerosis. Mult. Scler. Relat. Disord. 2023, 72, 104605. [Google Scholar] [CrossRef] [PubMed]
  51. Shahi, S.K.; Ghimire, S.; Lehman, P.; Mangalam, A.K. Obesity induced gut dysbiosis contributes to disease severity in an animal model of multiple sclerosis. Front. Immunol. 2022, 13, 966417. [Google Scholar] [CrossRef]
  52. Timmermans, S.; Bogie, J.F.; Vanmierlo, T.; Lütjohann, D.; Stinissen, P.; Hellings, N.; Hendriks, J.J.A. High fat diet exacerbates neuroinflammation in an animal model of multiple sclerosis by activation of the Renin Angiotensin system. J. Neuroimmune Pharmacol. 2014, 9, 209–217. [Google Scholar] [CrossRef]
  53. Asgharzadeh, V.; Rezaei, S.A.S.; Poor, B.M.; Asgharzadeh, M.; Nobari, H.J.; Taghinejad, Z.; Kazemi, A.; Rashedi, J. The Association between Diet and Multiple Sclerosis. Endocr. Metab. Immune Disord. Drug Targets 2024, 24, 909–917. [Google Scholar] [CrossRef] [PubMed]
  54. Coughlan, K.S.; Halang, L.; Woods, I.; Prehn, J.H. A high-fat jelly diet restores bioenergetic balance and extends lifespan in the presence of motor dysfunction and lumbar spinal cord motor neuron loss in TDP-43A315T mutant C57BL6/J mice. Dis. Models Mech. 2016, 9, 1029–1037. [Google Scholar] [CrossRef]
  55. Wang, S.; Yuan, T.; Yang, H.; Zhou, X.; Cao, J. Effect of complete high-caloric nutrition on the nutritional status and survival rate of amyotrophic lateral sclerosis patients after gastrostomy. Am. J. Transl. Res. 2022, 14, 7842–7851. [Google Scholar] [PubMed]
  56. Janse van Mantgem, M.R.; van Rheenen, W.; Hackeng, A.V.; van Es, M.A.; Veldink, J.H.; van den Berg, L.H.; van Eijk, R.P.A. Association Between Serum Lipids and Survival in Patients with Amyotrophic Lateral Sclerosis: A Meta-Analysis and Population-Based Study. Neurology 2023, 100, e1062–e1071. [Google Scholar] [CrossRef] [PubMed]
  57. Dorst, J.; Doenz, J.; Kandler, K.; Dreyhaupt, J.; Tumani, H.; Witzel, S.; Schuster, J.; Ludolph, A.C. Fat-rich versus carbohydrate-rich nutrition in ALS: A randomised controlled study. J. Neurol. Neurosurg. Psychiatry 2022, 93, 298–302. [Google Scholar] [CrossRef]
  58. Prado, M.B., Jr.; Pedro, K.M.; Adiao, K.J.B. Efficacy, safety and tolerability of high caloric diet in amyotrophic lateral sclerosis patients: A systematic review and meta-analysis. Rev. Neurol. 2023, 179, 1008–1019. [Google Scholar] [CrossRef] [PubMed]
  59. Villa, A.T.; Tu, B.H.; Titcomb, T.J.; Saxby, S.M.; Shemirani, F.; Ten Eyck, P.; Rubenstein, L.M.; Snetselaar, L.G.; Wahls, T.L. Association between improved metabolic risk factors and perceived fatigue during dietary intervention trial in relapsing-remitting multiple sclerosis: A secondary analysis of the WAVES trial. Front. Neurol. 2023, 13, 1022728. [Google Scholar] [CrossRef]
  60. Dean, Z.; Penesova, A.; Radikova, Z.; Dean, Z.; Kollar, B. Pilot study of longterm low fat diet in relapsing-remitting multiple sclerosis. Neuro Endocrinol. Lett. 2022, 43, 135–139. [Google Scholar] [PubMed]
  61. Harirchian, M.H.; Karimi, E.; Bitarafan, S. Diet and disease-related outcomes in multiple sclerosis: A systematic review of clinical trials. Curr. J. Neurol. 2022, 21, 52–63. [Google Scholar] [CrossRef] [PubMed]
  62. Parks, N.E.; Jackson-Tarlton, C.S.; Vacchi, L.; Merdad, R.; Johnston, B.C. Dietary interventions for multiple sclerosis-related outcomes. Cochrane Database Syst. Rev. 2020, 5, CD004192. [Google Scholar]
  63. Snetselaar, L.G.; Cheek, J.J.; Fox, S.S.; Healy, H.S.; Schweizer, M.L.; Bao, W.; Kamholz, J.; Titcomb, T.J. Efficacy of Diet on Fatigue and Quality of Life in Multiple Sclerosis: A Systematic Review and Network Meta-analysis of Randomized Trials. Neurology 2023, 100, e357–e366. [Google Scholar] [CrossRef] [PubMed]
  64. Zielińska, M.; Michońska, I. Effectiveness of various diet patterns among patients with multiple sclerosis. Postep. Psychiatr. Neurol. 2023, 32, 49–58. [Google Scholar] [CrossRef] [PubMed]
  65. Garodia, P.; Hegde, M.; Kunnumakkara, A.B.; Aggarwal, B.B. Curcumin, inflammation, and neurological disorders: How are they linked? Integr. Med. Res. 2023, 12, 100968. [Google Scholar] [CrossRef] [PubMed]
  66. Ahmadi, M.; Agah, E.; Nafissi, S.; Jaafari, M.R.; Harirchian, M.H.; Sarraf, P.; Faghihi-Kashani, S.; Hosseini, S.J.; Ghoreishi, A.; Aghamollaii, V.; et al. Safety and Efficacy of Nanocurcumin as Add-On Therapy to Riluzole in Patients With Amyotrophic Lateral Sclerosis: A Pilot Randomized Clinical Trial. Neurotherapeutics 2018, 15, 430–438. [Google Scholar] [CrossRef] [PubMed]
  67. Sathyabhama, M.; Priya Dharshini, L.C.; Karthikeyan, A.; Kalaiselvi, S.; Min, T. The Credible Role of Curcumin in Oxidative Stress-Mediated Mitochondrial Dysfunction in Mammals. Biomolecules 2022, 12, 1405. [Google Scholar] [CrossRef]
  68. Pastula, D.M.; Moore, D.H.; Bedlack, R.S. Creatine for amyotrophic lateral sclerosis/motor neuron disease. Cochrane Database Syst. Rev. 2012, 12, CD005225. [Google Scholar] [CrossRef]
  69. Ebrahimi, A.; Kamyab, A.; Hosseini, S.; Ebrahimi, S.; Ashkani-Esfahani, S. Involvement of Coenzyme Q10 in Various Neurodegenerative and Psychiatric Diseases. Biochem. Res. Int. 2023, 2023, 5510874. [Google Scholar] [CrossRef]
  70. Hathcock, J.N.; Shao, A. Risk assessment for coenzyme Q10 (Ubiquinone). Regul. Toxicol. Pharmacol. 2006, 45, 282–288. [Google Scholar] [CrossRef]
  71. Novak, V.; Rogelj, B.; Župunski, V. Therapeutic Potential of Polyphenols in Amyotrophic Lateral Sclerosis and Frontotemporal Dementia. Antioxidants 2021, 10, 1328. [Google Scholar] [CrossRef] [PubMed]
  72. Mancuso, R.; del Valle, J.; Modol, L.; Martinez, A.; Granado-Serrano, A.B.; Ramirez-Núñez, O.; Pallás, M.; Portero-Otin, M.; Osta, R.; Navarro, X. Resveratrol improves motoneuron function and extends survival in SOD1(G93A) ALS mice. Neurotherapeutics 2014, 11, 419–432. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Shared pathophysiological mechanisms of ALS and MS.
Figure 1. Shared pathophysiological mechanisms of ALS and MS.
Sclerosis 03 00001 g001
Table 1. Most relevant studies on the high-fat diet–MS association.
Table 1. Most relevant studies on the high-fat diet–MS association.
Author (Year)Cohort DetailsMeasured OutcomesLimitationsConclusions
Research in animal modelsDavanzo et al. (2023) [50]C57BL/6 J females randomized into two groups (a standard diet or a high-fat diet)Spinal cord lesions and pro-inflammatory biomarkers EAE animal model
No difference in innate or adaptive immune cell compartments
Spinal cord lesions in myelinated regions and BBB disruption
Higher levels of pro-inflammatory cells
Shahi et al. (2022) [51]19 HLA-DR3 transgenic mice randomized into three groups (prediet, high-fat diet, normal chow) Gut microbiota depletion
Microbiome analysis
Intestinal permeability and inflammatory mediator measurement
EAE animal modelMice on high-fat diet showed gut microbiota alterations, increased gut permeability, and systemic inflammation
Timmermans et al. (2014) [52]20 Female EAE mice randomized into two groups (normal rodent chow versus Western-type diet)Immunohistochemical staining and real-time PCR to determine immune cell infiltration and inflammatory mediatorsEAE animal modelActivation of the renin–angiotensin system, increased immune cell infiltration and inflammatory mediator production in high-fat diet-treated EAE mice
Research in humansAsgharzadeh et al. (2024) [53]467 MS patients, 260 controls,
age under 15
Azeri population
Dietary demographic questionnaireUse of questionnaires
Homogeneous small cohort
MS patients had a significantly higher consumption of high-fat foods
Table 2. Most relevant studies on the high-fat diet–ALS association.
Table 2. Most relevant studies on the high-fat diet–ALS association.
Author (Year)Cohort DetailsType of DietMeasured OutcomesLimitationsConclusions
Research in animal modelsCoughlan et al., 2016 [54]39 TDP-43A315T male mice randomized into three groupsHigh-fat jelly diet
low-fat jelly diet
standard pellet diet
Assessment of lifespan and disease progression in vivoNew animal model for the study of ALSHigh-fat diet prevented sudden death and extended survival in this animal model
Research in humansDorst et al., 2022 [57]64 Patients with possible, probable, or definite ALS randomized into four groupsHigh-caloric fatty supplements
ultra-high-caloric fatty supplements
ultra-high-caloric, carbohydrate-rich supplements
control group
Follow-up over four weeks for gastrointestinal side effects and weight modificationSmall and heterogeneous cohortDespite gastrointestinal side effects, a non-significant trend for weight gain was observed in the groups receiving high-caloric supplements
Table 3. Diet-based therapies in ALS and MS.
Table 3. Diet-based therapies in ALS and MS.
Targeted DisorderDietary InterventionFindingsEvidence/Limitations
Amyotrophic lateral sclerosisCurcuminAntioxidant and anti-inflammatory effects; potential synergy with RiluzoleMixed human trial results
Nausea and gastrointestinal side effects.
CreatineNeuroprotection in animal modelsLimited improvements in human trials
Further research required
CoQ10Antioxidant with mitochondrial benefitsSafe
Potential drug interactions and gastrointestinal side effects.
Phytochemicals (e.g., carotenoids, resveratrol, terpenoids)Antioxidative and anti-inflammatory effects; cognitive benefits (resveratrol) Preliminary evidence from in vitro and animal studies
Further human trials needed
Multiple sclerosisLow-saturated-fat dietsBenefits on weight, cholesterol levels, fatigue (Swank, Wahls diets) Small pilot studies
High dropout rate
Limited data on long-term adherence and efficacy
Plant-based dietsImproved fatigue and quality of life in MS patientsEvidence from systematic reviews
Variability in diet composition and patient adherence
AntioxidantsMixed results for neuroprotectionLimited large-scale evidence
Variable study designs and outcomes
Polyunsaturated fatty acids (PUFAs)Uncertain impact on relapse rates or disabilityMixed evidence
Further research required
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Schreiner, T.G.; Iacob, L.; Croitoru, C.G.; Hodorog, D.N.; Cuciureanu, D.I. High-Fat Diet—Shared Environmental Risk Factor for Amyotrophic Lateral Sclerosis and Multiple Sclerosis. Sclerosis 2025, 3, 1. https://doi.org/10.3390/sclerosis3010001

AMA Style

Schreiner TG, Iacob L, Croitoru CG, Hodorog DN, Cuciureanu DI. High-Fat Diet—Shared Environmental Risk Factor for Amyotrophic Lateral Sclerosis and Multiple Sclerosis. Sclerosis. 2025; 3(1):1. https://doi.org/10.3390/sclerosis3010001

Chicago/Turabian Style

Schreiner, Thomas Gabriel, Liviu Iacob, Cristina Georgiana Croitoru, Diana Nicoleta Hodorog, and Dan Iulian Cuciureanu. 2025. "High-Fat Diet—Shared Environmental Risk Factor for Amyotrophic Lateral Sclerosis and Multiple Sclerosis" Sclerosis 3, no. 1: 1. https://doi.org/10.3390/sclerosis3010001

APA Style

Schreiner, T. G., Iacob, L., Croitoru, C. G., Hodorog, D. N., & Cuciureanu, D. I. (2025). High-Fat Diet—Shared Environmental Risk Factor for Amyotrophic Lateral Sclerosis and Multiple Sclerosis. Sclerosis, 3(1), 1. https://doi.org/10.3390/sclerosis3010001

Article Metrics

Back to TopTop