Next Article in Journal
Bispecific Antibodies, Nanobodies and Extracellular Vesicles: Present and Future to Cancer Target Therapy
Previous Article in Journal
Ngn2-Induced Differentiation of the NG108-15 Cell Line Enhances Motor Neuronal Differentiation and Neuromuscular Junction Formation
Previous Article in Special Issue
Mitochondrial Transplantation in Animal Models of Psychiatric Disorders: A Novel Approach to Psychiatric Treatment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

From Obesity to Mitochondrial Dysfunction in Peripheral Tissues and in the Central Nervous System

1
Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
2
Department of Biology, University of Naples Federico II, 80126 Naples, Italy
3
Department of Human Sciences and Quality of Life Promotion, San Raffaele Telematic University, 00166 Rome, Italy
4
Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy
5
Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
6
Department of Biology, Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
7
Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy
8
Clinical Nutrition Unit, Madonna Delle Grazie Hospital, 75100 Matera, Italy
9
Unit of Dietetics and Clinical Nutrition, Department of Innovation, Experimentation and Clinical Research, S. Maria Alle Scotte Hospital, University of Siena, 53100 Siena, Italy
10
Department of Pharmaceutical Sciences, University of Perugia, 06126 Perugia, Italy
*
Authors to whom correspondence should be addressed.
These authors contributed equally to the work.
Biomolecules 2025, 15(5), 638; https://doi.org/10.3390/biom15050638
Submission received: 15 March 2025 / Revised: 25 April 2025 / Accepted: 26 April 2025 / Published: 29 April 2025
(This article belongs to the Special Issue Mitochondria and Central Nervous System Disorders: 3rd Edition)

Abstract

:
Obesity is a condition of chronic low-grade inflammation affecting peripheral organs of the body, as well as the central nervous system. The adipose tissue dysfunction occurring under conditions of obesity is a key factor in the onset and progression of a variety of diseases, including neurodegenerative disorders. Mitochondria, key organelles in the production of cellular energy, play an important role in this tissue dysfunction. Numerous studies highlight the close link between obesity and adipocyte mitochondrial dysfunction, resulting in excessive ROS production and adipose tissue inflammation. This inflammation is transmitted systemically, leading to metabolic disorders that also impact the central nervous system, where pro-inflammatory cytokines impair mitochondrial and cellular functions in different areas of the brain, leading to neurodegenerative diseases. To date, several bioactive compounds are able to prevent and/or slow down neurogenerative processes by acting on mitochondrial functions. Among these, some molecules present in the Mediterranean diet, such as polyphenols, carotenoids, and omega-3 PUFAs, exert a protective action due to their antioxidant and anti-inflammatory ability. The aim of this review is to provide an overview of the involvement of adipose tissue dysfunction in the development of neurodegenerative diseases including Alzheimer’s disease, Parkinson’s disease, and multiple sclerosis, emphasizing the central role played by mitochondria, the main actors in the cross-talk between adipose tissue and the central nervous system.

Graphical Abstract

1. Introduction

Obesity, one of the most significant public health concerns worldwide [1], is a multifactorial pathology characterized by excessive accumulation of body fat [2], which develops when energy intake exceeds energy expenditure for metabolic and physical activity. The World Health Organization reported that worldwide obesity affects more than 1.9 billion adults, representing 30% of the global population [3]. Caused by genetic, behavioral, societal, and environmental factors, obesity is a strong risk factor for the onset of chronic non-communicable diseases such as type 2 diabetes, cardiovascular illness, various types of cancer, musculoskeletal disorders, and cognitive decline [2].
Obesity, characterized by chronic low-grade inflammation, has been linked to neurodegenerative diseases such as multiple sclerosis (MS), Alzheimer’s disease (AD), and Parkinson’s disease (PD) [4]. Under conditions of obesity, adipose tissue releases pro-inflammatory adipokines/cytokines into the bloodstream [5]. Several pieces of evidence indicate a link between adipose tissue dysfunction, blood–brain barrier (BBB) alteration, neuronal dysfunction, neuroinflammation, and cognitive decline processes. Impairment of BBB integrity leads to increased CNS vulnerability to external factors. In obesity, peripheral inflammatory cytokines invade the CNS, resulting in reduced synaptic plasticity and neurogenesis and promoting the development of neurodegenerative diseases [6]. In this context, mitochondria, organelles that are the main energy source for the cell, play an important role. In fact, numerous studies indicate their involvement in the dysfunction of both adipose tissue and brain tissue in obesity [7]. The aim of this review is to provide an overview of the involvement of adipose tissue dysfunction in the development of some neurodegenerative diseases, such as MS, AD, and PD, under obesity conditions, emphasizing the central role played by mitochondrial dysfunction. In addition, we describe the effects of some bioactive molecules involved in restoring and improving impaired mitochondrial function in neurodegenerative disorders (NDDs).

2. Obesity and Inflammation

Obesity, considered as a low-grade chronic inflammation, displays an increased level of inflammatory markers and infiltration of macrophages into white adipose tissue [8]. In obesity, macrophages infiltrating adipose tissue form corona-like structures surrounding adipocytes, leading to the overproduction of adipokines, which comprise pro-inflammatory mediators, such as interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α) [9].
Lipid accumulation causes hypertrophy of adipocytes, inducing activation of pro-inflammatory pathways in these cells, such as nuclear factor kappa B (NFκB) [8,10] which is involved in increased production of TNF-α and other pro-inflammatory adipokines. TNF-α is a pro-inflammatory cytokine produced mainly by macrophages and appears to be an important factor in the deregulation of adipokines in adipocytes. Furthermore, it can induce lipolysis, leading to the release of free fatty acids (FFAs). The released fatty acids, in turn, bind to Toll-like receptor 4 (TLR4), which is located on the surface of both adipocytes and macrophages. The binding of fatty acids to TLR4 receptors further activates the NFκB pathway, increasing the production of pro-inflammatory adipokines [11]. Chronic low-grade inflammation in adipose tissue is transmitted systemically, leading to the onset and progression of metabolic disorders [12]. Numerous studies have demonstrated that obesity and related metabolic disorders are conditions that affect not only peripheral tissues but also the central nervous system (CNS) [13].
Adipose tissue dysfunction was correlated with impaired brain metabolism, neuroinflammation, neuronal dysfunction, and cognitive decline [14]. Excessive saturated fat intake and obesity have been shown to reduce the integrity of the blood–brain barrier (BBB), causing brain injury [15]. Specifically, studies in rats fed a Western diet high in saturated fat and sugar showed an increased BBB permeability and increased recruitment of monocytes into the CNS [6]. The structural integrity of the BBB is one of the most critical factors in maintaining brain homeostasis and is regulated by intricate interactions among various cell types, such as endothelial cells, pericytes, and astrocytes [16]. When the BBB is damaged, peripheral inflammatory cytokines and leukocytes migrate into the brain, resulting in reduced synaptic plasticity and impaired neurogenesis [17]. Thus, in obesity, peripheral immune cells can be recruited into the CNS like macrophages in white adipose tissue. This contributes to the activation of microglia, astrocytes, and endothelial cells, resulting in the secretion of pro-inflammatory cytokines such as TNF-α and interleukin-1 beta (IL-1β), which play a key role in the neuroinflammatory process [18].
Saturated fatty acids (SFAs) have been shown activate microglia and trigger the release of pro-inflammatory mediators that can lead to neuronal death [19]. Fatty acids can cross the blood–brain barrier and therefore brain fatty acid levels could be influenced by their presence in the periphery. Therefore, increased levels of circulating fatty acids, as observed in obesity, cause increased brain uptake of SFAs from plasma across the blood–brain barrier, resulting in microglia activation [19]. In vitro studies suggest that SFAs activate microglia to a pro-inflammatory state, increasing the secretion of pro-inflammatory cytokines via Toll-like receptor (TLR)4/NFκB signaling [20]. In vivo experiments have also demonstrated that SFAs activate TLR2 and TLR4 in the rodent hypothalamus, prompting the production of pro-inflammatory cytokines [21]. Furthermore, a high-fat diet compromised neurogenesis in the rodent hippocampus, a brain structure related to cognition, memory, learning, and emotions [22], enhanced lipid peroxidation, and reduced levels of brain-derived neurotrophic factor (BDNF). A growing number of studies have shown that different brain regions, such as the cerebral cortex, hypothalamus, and hippocampus, are subjected to obesity-induced neuroinflammation, which leads to NDDs such as AD [23], PD [24], and MS [25].

3. Obesity and Neurodegenerative Disorders

A growing number of data have recently shown that obesity is related to an increased risk of developing neurodegenerative diseases such as AD, PD, and MS, as well as neurological and neurovascular impairments [4,6]. In obese individuals, the risk of developing AD and PD is double that of normal-weight individuals [26,27]. In addition, it was observed that obesity in early childhood and adolescence is a significant risk factor for susceptibility to NDDs [28]. A post-mortem study showed that elderly and morbidly obese individuals have elevated levels of NDD markers [29]. Higher body mass index (BMI) in middle age has been found to predispose to a greater risk of neurological impairment in later life [30].

3.1. Alzheimer’s Disease

AD is the most common form of dementia, contributing 60–70% of cases, with the prevalence in women 1.17 times higher than in men [31]. Dementia is currently the seventh leading cause of death and a major cause of disability and dependency in older people worldwide.
AD is an NDD, characterized by progressive neuronal and synaptic loss and biochemical abnormalities, including the deposition of extracellular amyloid beta (Aβ) plaques, tau hyperphosphorylation, and mitochondrial dysfunction, mainly in the cerebral cortex, with a bigger impact on the hippocampus [32]. The initial symptoms of the disease are memory loss, which then advances into various cognitive impairments and eventually leads to death. Several studies reported that obesity has an influence on Aβ deposition. Diet-induced obesity (DIO) is consistently linked to an increase in cerebral Aβ accumulation [33]. Long-term high-fat diet (HFD) consumption in rodents has been shown to result in increased levels of Aβ precursor protein (APP) in both the hippocampus and adipose tissue, combined with an increased inflammatory state. Aβ peptide has been shown to exert functional effects on adipose tissue, leading to increased release of FFAs and pro-inflammatory adipokines [34].
Likewise, a marked association between weight reduction and lower levels of Aβ plaques in the brain has been reported using various dietary regimens, including ketogenic and low-calorie diets [35,36] in different APP transgenic strains, demonstrating that body weight, diet, and obesity have a significant impact on Aβ pathology.
Furthermore, studies have shown that obesity modulates neurofibrillary tangles (NFTs) produced by hyperphosphorylated tau, the microtubule-associated protein [37]. In AD transgenic mice, it was observed that DIO increased Aβ levels and tau phosphorylation. Specifically, an HFD causes Aβ accumulation and tau hyperphosphorylation in the frontal cortex of the 3 × Tg AD mouse model [38].
In addition, in two AD mouse models (3 × TgAD and 5 × FAD), HFD administration is associated with decreased synaptic contacts, signs of cognitive impairment, and oxidative stress in the hippocampus [39].

3.2. Parkinson’s Disease

Parkinson’s disease (PD) is the second most common neurodegenerative disease of the CNS after AD. The incidence of PD ranges from 5/100,000 to more than 35/100,000 new cases per year. From the sixth to the ninth decade of life, the incidence increases 5- to 10-fold. PD is characterized by symptoms such as tremor, bradykinesia, muscle rigidity, and impaired posture and balance [40].
A distinctive sign of PD is the degeneration of dopaminergic neurons of the nigrostriatal pathway and the development of intracellular protein aggregates, mainly composed of alpha-synuclein (αSYN), named Lewy bodies, within the neurons [41]. It has been suggested that some of the factors responsible for the degeneration of the dopaminergic system in the CNS, leading to PD, are mitochondrial dysfunction, oxidative stress, and neuroinflammation [42]. Accumulating epidemiological evidence indicates that obesity is a primary risk factor for the development of PD [43].
In obese individuals, adipose tissue has been observed to yield inflammatory adipokines that potentially accelerate disease progression [44]. Evidence shows that saturated FAs are implicated in the pathogenesis of PD, potentially influencing αSYN aggregation, destruction of dopaminergic neurons, oxidative stress, and cytokine production [45]. Studies found that rodents fed with a 45–60% fat diet for 3 to 5 months showed a significant alteration in dopamine receptor expression and dopamine release in the midbrain, with dopamine uptake slowing as of the third month of HFD intake [46]. Furthermore, dopamine receptors and transporters decreased by 42% and 30%, respectively, in rats fed with a high-fat diet for eight weeks [47]. Another study revealed an alteration of dopaminergic neurotransmission after only two weeks of HFD consumption (60%) [48].

3.3. Multiple Sclerosis

MS is the most prevalent chronic inflammatory illness of the CNS, affecting over 2.8 million people worldwide. It develops between the ages of 20 and 40 and is more common in young adults and especially women [49].
MS, which affects the brain and spinal cord, is characterized by demyelinating lesions in the white matter of the CNS with axonal degeneration. These lesions cause typical but also unspecific neurological symptoms: difficulties with vision, movement, or balance, leading to disability and reduced quality of life [50]. Recent reports have highlighted that the interaction of environmental and lifestyle risk factors with an individual and susceptible genetic background is implicated in MS development.
In recent decades, risk factors such as obesity, type 2 diabetes, and dyslipidemia have been associated with the onset and progression of MS. Indeed, recent reports showed a correlation between childhood and adolescent obesity and the risk of developing MS [51]. In addition, a study of 8983 patients with MS showed that 31.3% of patients were overweight and 25% were obese [52,53], demonstrating a high incidence of overweight and obesity in the MS population. Furthermore, it has been shown that lipid metabolism disorder is more common in MS subjects compared to normal subjects, with higher levels of oxidized low-density lipoprotein and small high-density lipoproteins [54].
Recently, leptin, an adipokine produced by adipocytes in proportion to the mass of adipose tissue, has been suggested to play a relevant role in the regulation of autoimmune and inflammatory processes and MS disease progression [55]. Leptin is increased in MS patients and adversely correlated with Foxp3 gene expression in PBMCs, which plays a role in suppressing immune function, and is positively related to TNF-α, IL-1β, and hsCRP levels [56]. High levels of leptin may create a positive feedback loop leading to MS progression, as higher levels of this adipokine cause reduced Foxp3 expression and increased production of pro-inflammatory cytokines. These cytokines further reduce Foxp3, which in turn increases leptin and pro-inflammatory cytokines [56].
Recent studies have highlighted that caloric restriction helps to reduce serum leptin levels, and this may increase the survival and lifespan of animal models of inflammation, including those that resemble human MS [57]. Interestingly, calorie restriction reduces inflammation, demyelination, and axon injury without impairing the function of the immune system response [57].

4. Obesity, Mitochondrial Dysfunction, and Neurodegenerative Disorders

4.1. Obesity and Adipose Tissue Mitochondrial Dysfunction

Mitochondria are double membrane organelles important to the production of ATP in the cell and involved in the adaptation of energy demand, thus playing a relevant role in bioenergetic metabolism. These organelles also represent the main site of cellular production of reactive oxygen species (ROS). Mitochondrial function is closely linked to ROS levels. Under normal conditions, the generated ROS are seized by antioxidant enzymes such as superoxide dismutase, glutathione peroxidase, and catalase [58]. Under conditions of disease or high energy demand, ROS production exceeds the antioxidant capacity of the cell, resulting in oxidative damage. Excessive ROS production is known to damage mtDNA, leading to mitochondrial dysfunction [59]. In diet-induced or genetic mouse models of obesity, decreased OXPHOS capacity and mitochondrial biogenesis were observed in white adipocytes [60,61]. In addition, the number of gene transcripts encoding mitochondrial proteins decreased in obese mice [62], while a decline in mitochondrial mtDNA was observed in obese subjects [63]. Furthermore, both the activity of OXPHOS complexes I to IV and mitochondria’s membrane potential were downregulated in the subcutaneous adipose tissue of obese individuals and in isolated adipocyte mitochondria [64].
Numerous studies have indicated that mitochondrial dysfunction anticipates inflammation in adipose tissue, leading to elevated TNF-α levels, ER stress, and increased ROS in mouse 3T3-L1 pre-adipocytes [65]. Indeed, mitochondrial dysfunction leads to increased ROS generation which activates nuclear factor kappa B, resulting in increased expression of pro-inflammatory cytokines [66] and immune cell infiltration [67,68]. Adipocytes are known to play a significant role in the production of monocyte chemoattractant protein-1 (MCP-1), chemokines that regulate monocytes/macrophage migration and infiltration [10]. It has been observed that mitochondrial dysfunction in adipocytes causes increased expression of MCP-1 [69], which increases macrophage chemotaxis into adipose tissue.
Chronic low-grade inflammation caused by mitochondrial dysfunction in adipose tissue is transmitted systemically, leading to metabolic disorders that also impact the CNS, resulting in the initiation and progression of neurodegenerative diseases (Figure 1).
Indeed, numerous studies have demonstrated the relevant role of mitochondria in the development and progression of obesity-induced metabolic disorders in different organs and tissues [61,70,71].

4.2. Obesity and Brain Mitochondrial Dysfunction

The CNS, representing only 2% of total body weight, consumes, at rest, about 20% of the inhaled oxygen. Neurons, which are responsible for about 75–80% of the brain energy production, are critically dependent on mitochondrial energy metabolism [72]. It was estimated that there are about two million mitochondria in a neuron [73]. Accordingly, multiple neural functions are supported by mitochondrial ATP, including ion buffering, synaptic vesicle recycling, neurotransmitter synthesis, axonal transport, and the assembly of the actin cytoskeleton [74]. Mitochondria located in the synaptic regions of the neuron play a crucial role in providing ATP for synaptic functions, such as supporting the local system of protein synthesis necessary for synaptic plasticity [75]. Synaptic plasticity is the ability of the nervous system to rapidly adapt to environmental changes, through modification in the number and strength of synaptic contacts during development and in adulthood [75,76,77]. The axonal terminals and dendritic spines are highly plastic sites with enormous energy demands, characterized by a great number of mitochondria. Inhibition of mitochondrial activity has been shown to result in altered synaptic potentiation and compromised neurotransmission, while stimulation of mitochondrial respiration has led to an increase in synaptic density [78]. A reduction in the number of mitochondria is associated with the loss of synapses and spines, as well as a decrease in dendritic length in neuronal cultures [79]. Thus, it is possible to conclude that synaptic functionality and plasticity phenomena depend on mitochondrial activity. Accordingly, mitochondrial dysfunction is directly linked to compromised synaptic plasticity and neuroinflammation [80,81], which are common features of several neuropathologies [82,83]. Nonetheless, it is not yet clear whether mitochondrial dysfunction is a cause or a consequence of inflammatory processes, triggering metabolic adaptations that may lead to protective or detrimental pathways.
Numerous findings correlate mitochondrial dysfunction with different neurodegenerative diseases, such as AD, PD, and MS, but also progressive myoclonic epilepsy type 1 and Friedreich’s ataxia [84,85].
More evidence underscores the impact of dietary fat on brain function and cognitive deficits, indicating that long-term intake of HFDs leads to changes in brain mitochondrial function [51,86]. An HFD has been observed to alter mitochondrial function, leading to reduced oxidative capacity in the cerebral cortex and synaptosomal fraction. Recently, alterations in synaptic plasticity and energy metabolism have been demonstrated in an animal model of diet-induced obesity [80]. An HFD was found to reduce uncoupling in brain mitochondria, resulting in increased production of ROS [80]. Mitochondrial uncoupling is a natural physiological mechanism to limit excessive ROS production. Mitochondria generate ATP via oxidative phosphorylation, but part of the energy produced by electron transport is uncoupled from ATP synthesis due to the proton leak across the inner mitochondrial membrane. This uncoupling allows the mitochondrial membrane potential to be kept below the critical threshold for ROS production. Therefore, ROS generation is minimized by the decrease in mitochondrial coupling [87]. In animal models of diet-induced obesity, increased levels of oxidative stress markers such as MDA and ROS were observed in the hypothalamic region [88] and increased mitochondrial peroxide yield in the hippocampus [89].
In addition, decreased expression of proteins involved in mitochondrial activity was observed in the brains of obese mice, specifically mitochondrial cytochrome c oxidase (COX) subunit 7B (components of complex I and complex IV of the respiratory chain) [90].
In conclusion, loss of neuronal mitochondrial function at the level of synaptic regions impairs neuronal plasticity, as they contribute to the blockage of neurotransmission and cognitive dysfunction associated with neurodegenerative diseases [91]. Evidence correlating mitochondrial dysfunction with major neurodegenerative diseases is given below.

4.3. Mitochondrial Dysfunction in Alzheimer’s Disease

AD is a complex pathology in which mitochondrial dysfunction plays a prominent role. Indeed, in AD, mitochondrial dysfunction is responsible for disrupting the normal functioning of synapses, resulting in a reduced neurotransmission and weakened synaptic connections. This impairment contributes to cognitive decline and memory impairment in AD [92]. Accordingly, AD patients display decreased cytochrome c oxidase activity and several deficits in the activity of other mitochondrial enzymes. Moreover, AD neurons show reduced ATP levels and increased ROS levels [93].
A mouse model of AD, overexpressing mutant APP in the brain, shows several alterations at the mitochondrial level. For instance, impaired mitochondrial morphology, increased ROS production, and diminished membrane potential and respiration were observed [94]. It has been proposed that Aβ interferes with mitochondrial function, causing the metabolic deficiencies and neurological dysfunction observed in the brains of AD patients [95]. Moreover, in AD brain and neuronal cultures, Aβ has been shown to disrupt the endoplasmic reticulum–mitochondria connection, resulting in altered mitochondrial morphology, motility, bioenergetics, autophagy, apoptosis, and Ca2+ signaling [96]. In addition, it was observed that mitochondria represent the intracellular site of Aβ accumulation in hippocampal regions of APP transgenic mice [97].
It is well known that excessive/abnormal phosphorylation of tau leads to the formation of insoluble fibers and NFTs, a pathological hallmark of AD, which impair cell function and axonal transport [98]. Studies suggest a nexus between tau and mitochondrial dysfunction. Indeed, an overexpression of tau was observed to induce changes in mitochondrial morphology, mitochondrial transport, and decreased complex I and ATP activity [99]. It has been proposed that tau can be localized in the inner mitochondrial sub-compartments, located between the outer mitochondrial membrane and the inner mitochondrial space, but not in the mitochondrial matrix and could affect the development of contact sites between the ER and the mitochondria [100].
Overexpressed and hyperphosphorylated tau has been shown to impair the localization and distribution of mitochondria, causing defects in axonal function and losses in synapses [101] in several cellular and mouse models of AD. Similarly, alterations in mitochondrial localization have been observed in human AD brains, confirming the link between tau accumulation and mitochondrial translocation [102]. Furthermore, it has been widely observed that the dynamic balance between mitochondrial fission and fusion is disrupted in AD, resulting in a shift toward immoderate fission [103]. Specifically, an abnormal interaction between hyperphosphorylated tau and Drp1, a protein that regulates mitochondrial fission, was observed in brain tissue from 3 ×Tg-AD mice and in brain tissue from AD patients, resulting in excessive mitochondrial fission that induces mitochondrial dysfunction and synaptic degeneration [104].
Long-term mitochondrial fission has also been evidenced in in vitro models of AD. Indeed, overexpression of APP or treatment with Aβ causes severe fragmentation and altered allocation of mitochondria, which likely triggers Aβ-induced synaptic defects in neuronal cultures [105].

4.4. Mitochondrial Dysfunction in Parkinson’s Disease

Parkinson’s disease is another neurodegenerative disease in which mitochondrial dysfunction plays a key role and is characterized by the loss of dopaminergic neurons and intracellular protein aggregates, mainly composed of αSYN “Lewy bodies”. The Lewy bodies, a feature of PD, have been demonstrated to accumulate in organelles including mitochondria and both mitochondrial dysfunction and oxidative stress have been linked to PD [106]. Both PD patients and animal models of PD display decreased activities of complex I (CI) of the mitochondrial respiratory chain. Furthermore, partial inhibition of complex I in rat synapses has been shown to increase mitochondrial ROS production [107]. Inhibition of complex I can interfere with energy/ATP generation, causing partial neuronal depolarization attributed to a decrease in Na+/K+-ATPase activity and increased ROS production [108]. In turn, ROS inhibit complex I, creating positive feedback that generates more ROS, increasing oxidative stress and depleting ATP. This alteration of nerve terminal mitochondria has been proposed as a mechanism to explain the neuronal loss in the nigrostriatal pathway that characterizes PD [109]. Therefore, mitochondrial dysfunction has been identified as playing a crucial role in the development of PD.
Neuroinflammation contributes to increased oxidative stress and dopaminergic neuronal loss in the brain of PD patients. Excessive ROS production can cause transcription errors that lead to dysfunction in the expression of several proteins, such as C-terminal αSYN, parkin, and ubiquitin hydrolase, which are directly linked to PD [110]. Mutations in several PARK genes affect mitochondrial function in PD development. Early onset autosomal recessive PD is most commonly caused by mutations in the genes encoding PINK1 (PARK6) and Parkin (PARK2). PINK1 and Parkin serve key roles in mitochondrial quality control mechanisms and the response to mitochondrial damage [111]. PINK1/Parkin has a significant role in the control of mitochondrial fission and fusion and in the induction of mitochondrial biogenesis [112]. The use of Parkin-KO mice in experimental studies showed a decreased mitochondrial respiratory capacity [113]. PINK1-KO mice also showed defects in CI function, alterations in mitochondrial membrane potential, and an increase in mitochondrial fission [114].
Defects in mitochondrial biogenesis were also observed in Parkin-deficient human dopaminergic neurons. Both Parkin and PINK1 mutation or loss in human SH-SY5Y cells result in exacerbated mitochondrial fragmentation mediated by Drp1 [115]. Mutations in the gene encoding for DJ1 (PARK7) are also implicated in PD. Subjects with DJ1 mutations are characterized by early onset and slow progression of Parkinsonism [116]. DJ1 depletion leads to impaired mitochondrial respiration, elevated intracellular ROS levels, impaired mitochondrial membrane potential, and altered mitochondrial morphology [117]. Experimental evidence from isolated mitochondria [118] and rodent tissue [119] underscores how αSYN is a major cause of mitochondrial dysfunction in PD. αSYN has been shown to be a regulator of mitochondria oxidative phosphorylation by interacting with complex I and complex V [120]. Accumulation of oligomers of αSYN or misfolded αSYN in mitochondria reduces complex I activity, ATP synthesis, and organelle biogenesis and increases ROS production, leading to an overall alteration in mitochondrial function and dynamics [121].
Conversely, monomeric αSYN is a physiological regulator of mitochondrial bioenergetics through its ability to interact with ATP synthase and increase its efficiency. Indeed, αSYN knockout mice exhibited lower ATP synthase efficiency, diminished levels of ATP, and an altered neuronal mitochondrial membrane structure and a deficiency of complex I [122].
It has been observed that the interaction of pathological oligomers of αSYN with the alpha subunit of ATP synthase may be mediating aSYN-induced mitochondrial dysfunction [123]. In addition, αSYN oligomers have been seen to interact with protein TOM20, an external mitochondrial membrane protein. As a result of this binding, mitochondrial proteins are compromised and the electron transport chain malfunctions, altering the mitochondrial membrane potential and leading to the accumulation of ROS [124].

4.5. Mitochondrial Dysfunction in Multiple Sclerosis

It is well known that the neurodegeneration observed in MS is caused by multiple factors, and mitochondrial dysfunction appears to play a central role, contributing to impaired myelination and axonal damage [125]. N-acetylaspartate is a mitochondrial metabolite and substrate for myelin production by oligodendrocytes (after decomposition into acetate and aspartate). Decreased N-acetylaspartate uptake from dysfunctional mitochondria lead to the low myelination observed in MS patients [126].
The reduction of myelin observed in MS is followed by an increase in the energy requirements of the axon to maintain its resting membrane potential [127]. Neurons and oligodendrocytes (OLs) in post-mortem MS tissues and animal models of MS have shown alteration of normal mitochondrial functions [127]. Accordingly, a decreased expression of mitochondrial complexes was observed in upper cortical motor neurons of MS patients [128]. Several studies revealed that inflammation-related mitochondrial damage in MS is linked to the blockade of the mitochondrial chain complex IV [129], a pivotal component of mitochondrial activity responsible for approximately 90% of total cellular oxygen consumption. An increased activity of complex IV has been demonstrated in chronic demyelinating lesions [130], suggesting that this complex may play a crucial role in counteracting the energetic consequences of inflammation-related axonal demyelination. Nitric oxide (NO), a mediator released during inflammatory demyelination, and related species could be detrimental to mitochondrial complex IV because they can compete with molecular oxygen (O2) in binding the functional site of COX, the main subunit of the complex, resulting in transient inhibition and reversible conduction block in axons [131]. Overproduction of NO is one of the features of neuroinflammation-related CNS diseases, including MS. The role of pro-inflammatory cytokines in NO production is significant in both the peripheral immune system and the CNS [132].
Furthermore, a decrease in PGC-1 levels and in the expression of various mitochondrial protein components in pyramidal neurons has been observed in patients with MS [133]. In addition, higher levels of a heat shock protein (mtHSP70), a marker of mitochondrial stress, were found in MS subjects [134]. Moreover, an enhanced mitophagy was detected in neurons of MS patients, although it is not clear whether this process is associated with pathogenesis or represents a compensatory mechanism [135].
In mitochondria-enriched fractions isolated from the brain cortex of post-mortem MS patients, a reduction in mitochondrial electron transport chain gene expression was observed but no deficiency in the number of mitochondria themselves [128]. In addition, complexes I and III were found to be decreased by 61 and 40%, respectively (compared to control), but there were no variations in complex IV [128]. Several studies have been conducted using animal models of MS, such as experimental autoimmune encephalomyelitis (EAE) mice and cuprizone (CPZ)-induced demyelination mouse models.
In animal models of MS, demyelination has been shown to have a direct impact on mitochondrial dynamics through changes in fission and fusion. Specifically, an increase in DRP1 expression was observed in the spike-injured spinal cords of EAE mice and in the corpus callosum of CPZ mice. While inhibiting Drp1 activation, a neuroprotective effect has been found in both EAE and CPZ [136]. In addition, an enhanced Mfn2 expression was observed in proteolipid (PLP)4e mice, a demyelinating mouse model containing extra copies of myelin genes [137].

5. The Role of Bioactive Compounds in Obesity and Neurodegenerative Disorders: Focus on Mitochondrial Function

To date, many studies conducted both in vivo and in vitro have demonstrated the important role played by some biomolecules, such as polyphenols, carotenoids, fatty acids, and endocannabinoid-like compounds, in the prevention of or recovery from obesity and NDDs (Figure 2). The studies reported below illustrate the biological effects of the administration of pure molecules in vivo or in vitro experimental models.

5.1. Polyphenols

Polyphenols, classified as secondary plant metabolites, are known for their antioxidant properties and cytoprotective and anti-inflammatory effects [138].
Resveratrol (RES, 3, 5, 4′-trihydroxystilbene) and curcumin (CM) have been proven to have cytoprotective effects, preventing and/or minimizing cellular and mitochondrial damage. In addition, these compounds exhibited both anti-lipid effects and relevant neuroprotective activities, among many other beneficial properties. In general, the neuroprotective benefits of a diet rich in polyphenols have been studied and confirmed by numerous studies in cell and animal models [139].
RES is a phenolic micronutrient compound, which occurs naturally in blueberry, grapes, mulberry, and other fruits. The precise mechanism by which RES achieves its beneficial effects are not yet fully understood. Several recent pieces of evidence have demonstrated that it performs its action through mitochondria modulation, proving to be a substance capable of enhancing their functions [140]. The inhibitory effect of RES on adipogenesis has been examined by several studies conducted in vitro and in animal models. In pre-adipocyte cells incubated with increasing concentrations of RES (1, 10, and 25 µM) for 24 h, a decrease in triacylglycerol content and decreased expression of acetyl-CoA carboxylase (ACC), the first enzyme with a key role in the synthesis of long-chain saturated fatty acids, were found [141]. In an animal model of obese Zucker rats, a reduction in abdominal fat and ACC enzyme activities was found after administration of RES 10 mg/kg for 8 weeks [142]. RES would hinder the process of adipogenesis by activating the AMPK signaling pathway [143], an energy sensor responsible for metabolic modifications by phosphorylating downstream substrates, such as ACC. RES exert an anti-adipogenic effect by promoting the expression of genes involved in oxidative phosphorylation and mitochondrial biogenesis through stimulation of SIRT-1 deacetylase [144]. Moreover, studies have demonstrated the ability of RES to prevent cognitive impairment and neurodegeneration through its antioxidant and anti-inflammatory properties. RES enhances the level of cellular antioxidants, such as glutathione, superoxide dismutase, glutathione peroxidase, and catalase, and reduces lipid damage due to the peroxidation events [145] in brain tissue. RES has been shown to decrease superoxide radical synthesis by stimulating complex III activity and COXIV activity [146].
Reports suggest that RES can be used to treat and manage AD, exerting a protective action against Aβ-induced neuronal oxidative damage [147,148]. RES ameliorates motor dysfunction in the A53T αSYN mouse model of PD and reduces striatal αSYN levels, glial activation, and TNF-α and IL-1β levels in an MPTP mouse model of PD [149]. RES improves brain mitochondrial health by modulating the activation and expression of peroxisome proliferator-activated receptor coactivator-1α (PGC-1α), which is liable for mitochondrial biogenesis [149], boosting mitochondrial mass.
CM is a polyphenol derived from Curcuma longa L. and is known to exert multiple beneficial effects including weight loss and reduction in the incidence of obesity-related diseases [150]. CM inhibits adipocyte differentiation, promotes antioxidant activities, and prevents macrophage infiltration and activation of nuclear factor κB (NFκB) in adipose tissue [151]. Moreover, CM was observed to remarkably improve mitochondrial respiratory function in mature adipocytes, specifically accelerating mitochondrial basal respiration, ATP production, and expression of mitochondrial uncoupling protein 1 (UCP1). CM performs these actions via the regulation of peroxisome proliferator-activated receptor-γ (PPAR-γ) in 3T3-L1 adipocytes and in an obese rodent model [152]. In addition, studies have shown its beneficial therapeutic properties in CNS cells, particularly in attenuating mitochondrial dysfunction. CM has been shown to improve neuronal survival by reducing apoptosis, oxidative stress, and neuroinflammation via activation of the CREB-BDNF pathway [153]. CM has been observed to act by activating Nrf2 and Nrf2 target genes in primary astrocytes, thereby reducing intracellular ROS levels and attenuating oxidative damage and mitochondrial dysfunction. CM restores mitochondrial function through induction of the nuclear receptor PGC1α, showing itself as a hopeful agent that can delay brain aging and forestall mitochondrial damage [154].

5.2. Carotenoids

Carotenoids (CTs) are substances corresponding to a large family of C40 lipophilic pigments produced by photosynthetic organisms and some prokaryotes and fungi. They play an important role as antioxidants, primarily as lipid defenders against the damaging effects of oxidative stress.
CTs have been shown to have a significant impact on the treatment of obesity and neurodegenerative diseases [155]. They perform their function by maintaining mitochondrial function and integrity [156]. Studies have been conducted to examine the potential use of CTs in the managing of obesity. In particular, the effects of pure carotenoid supplementation on obesity have been investigated in two randomized double-blind placebo-controlled clinical trials. In the first, a reduction in BMI, waist-to-height ratio, and subcutaneous adipose tissue was observed in children who received a carotenoid mixture for 6 months [157]. In the second, a mixture of CTs was given to healthy overweight volunteers for 12 weeks, resulting in a reduction in total fat area and BMI compared with a placebo group [158]. CTs have been observed to act by inhibiting adipocyte differentiation by interfering with nuclear receptors like RAR, RXR, or PPAR [159]. Studies suggest that the brain is involved in body weight regulation under the influence of CTs. These compounds have been found in various regions of the adult brain [160]. Interestingly, it has been suggested that these compounds would cross the blood–brain barrier and affect neurons in the arcuate nucleus, influencing feeding behavior and preventing weight gain and adiposity [161].
CTs are considered beneficial in the treatment of NDDs, exhibiting anti-neuroinflammatory properties and a significant role in the prevention of PD and AD. Indeed, CT supplementation has been shown to be effective in hindering αSYN toxicity and protecting against the amyloid and tau diseases [155,162]. These beneficial effects are due to their ability to improve mitochondrial function. In support of this, it has been shown that deficiency of the enzyme β-carotene oxygenase 2 (BCO2), which is involved in the catalytic activities of CTs, leads to superoxide overproduction in mitochondria and decreases the level of mitochondrial superoxide dismutase, affecting mitochondrial respiratory complexes [163].
Lycopene (LYC) is the main carotenoid present in tomatoes and tomato products, and its anti-obesity effect has been shown in mice fed a high-fat diet, whose adiposity was reduced after supplementation. LYC could reduce body weight gain by increasing energy expenditure. Indeed, LYC was found to increase mitochondrial biogenesis and function in obese mice by increasing basal mitochondrial respiratory capacity and ATP-linked respiration. Furthermore, LYC increases PGC-1α and UCP1 expression in mature adipocytes and adipose tissue [164]. LYC would exert its action by downregulating lipogenesis genes and upregulating those related to lipidolysis, such as thermogenic and mitochondrial functional genes [165]. Recent advances have demonstrated a neuroprotective effect of LYC. In particular, it improves cognitive abilities and alleviates memory impairments in tau transgenic mice, an animal model of AD [166]. In addition, this compound decreases the secretion of Aβ, downregulates amyloid precursor protein (APP) levels, and decreases tau hyperphosphorylation in the brain [166]. LYC has been reported to restore mitochondrial dysfunction in AD and PD through its antioxidant and anti-inflammatory properties. Specifically, it can prevent Ab-induced cell damage by activating the PI3K/Akt/Nrf2 pathway [91,167,168].
Astaxanthin (ASX), also called 3,3′-dihydroxy-β, β′-carotene-4,4′-dione, is a carotenoid with a bright orange to red color found in marine animals such as shrimp, lobster, crab, and salmon and some other organisms such as algae. In terms of antioxidant activity, ASX is ten times more potent than other CTs and 100 times more potent than α-tocopherol [169]. ASX has been shown to improve obesity and its associated conditions. Indeed, its supplementation and physical training over a 12-week period in a group of obese men led to a reduction in adiponectin levels and body weight and fat percentage and improved lipid and metabolic profiles. In addition, ASX has been observed to inhibit body weight gain and lipid accumulation and reduce the release of pro-inflammatory cytokines in mice fed a high-fat diet [170]. This is due to its ability to activate AMPK and upregulate the expression of transcription factors involved in mitochondrial remodeling and increased mitochondrial component of oxidative phosphorylation [171]. Furthermore, ASX has been reported to contribute to the enhancement and maintenance of mitochondrial activity, promote mitochondrial biogenesis, and inhibit mitochondrial fission/fragmentation by activating antioxidant and anti-inflammatory pathways in various cell lines [172]. ASX acts by inhibiting canonical NFκB signaling in response to oxidative stress. Specifically, it suppresses NFκB-mediated gene expression of pro-inflammatory cytokines such as IL-1β, IL-6, or TNF-α, thereby inhibiting the development of inflammation. Current studies show that ASX has a preventive effect on oxidative-stress-induced neurodegenerative pathological conditions by maintaining the integrity of the BBB and ameliorating neuroinflammation induced by the neurotoxin or stressful conditions such as glucose deprivation/reperfusion treatment in the rodent brain [173]. Specifically, ASX acts by preserving the stability of tight junctions of microvascular endothelial cells [174]. In a mouse model of AD, ASX in the ester form with docosahexaenoic acid decreases oxidative stress and inflammasome activation [175].

5.3. Fatty Acids

After adipose tissue, the brain is the organ with the highest amount of lipids, with approximately 50% of its dry weight being lipids. Lipids play a crucial role in the normal development and function of the CNS. The type of FAs incorporated into the phospholipids of brain cell membranes determines membrane structure, microdomain organization, and fluidity. Data have shown the impact of dietary lipids on the degree of lipid accumulation in the body, as well as the structure and functioning of the brain [176]. Accordingly, alterations in cholesterol metabolism have been implicated in the pathogenesis of several NDDs [177]. On the other hand, numerous studies clearly demonstrate that consumption of omega-3 polyunsaturated fatty acids (PUFAs), such as docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), attenuates diet-induced obesity by increasing mitochondrial lipid oxidation and reducing energy efficiency through modulation of the mitochondrial uncoupling [178,179]. In addition, omega-3 PUFAs, particularly DHA, can affect brain function [180]. Indeed, a positive association between high DHA consumption or high serum DHA levels and a reduced risk of developing AD was observed in a population of patients aged 65 to 94 years. In addition, DHA demonstrated a protective effect against typical AD pathological mechanisms [181]. Accordingly, recent prospective questionnaire-based studies have reported that a high intake of unsaturated fatty acids may protect against PD [182].
It is well known that PUFAs have important effects on phospholipid composition of mitochondrial membranes and on the functionality of these organelles, as shown in experiments conducted on animal models of aging, AD, and PD, which exhibited an improvement in brain mitochondrial function after treatment with PUFAs such as DHA [183,184]. Specifically, ω-3 PUFAs exert neuroprotective effects by counteracting oxidative stress and consequent alterations in the structure and function of mitochondrial transporters and enzymes involved in ATP production and glucose metabolism. Different studies confirm the beneficial effects of omega-3 PUFAs on inflammatory parameters by modulating mitochondrial respiratory parameters in the liver, skeletal muscle, and brain through supplementation intake in animal models [185,186,187].
Conjugated linoleic acids (CLAs) are a class of isomers of linoleic acid, known to exhibit important beneficial effects on human health. Several studies have shown their ability to counteract HFD-induced obesity [155,188,189]. CLAs have been observed to activate several nuclear receptors involved in the regulation of gene expression related to lipid metabolism. Specifically, CLAs decrease lipid synthesis, adipogenesis, and lipid storage in adipocytes and increase β-oxidation in skeletal muscle [190]. Furthermore, the trans-10, cis-12 isomers have been shown to influence body composition, decreasing adiposity by promoting the browning of adipocytes in mice [191]. In addition, the preventive effect of CLA supplementation against age-dependent neurodegeneration in the mouse model has been demonstrated, likely due to its effect in reducing oxidative stress and improving mitochondrial function, also noted in various organs and tissues [192,193]. This protective effect may occur through the activation of nuclear factor erythroid 2-related factor (Nrf2), which plays a key role in the preservation of redox homeostasis and mitochondrial function and preventing the inflammatory state [194].
Short-chain fatty acids (SCFAs, microbial metabolites such as butyrate, propionate, and acetate) have been shown to have many beneficial effects on human health, including an anti-obesity effect [195]. In fact, human studies have demonstrated that administration of SCFA to obese patients resulted in the increased production and secretion of the satiety-promoting hormone peptide YY (PYY) and glucagon-like peptide-1 (GLP-1), with consequent reduction in adiposity [196]. Furthermore, studies in animal models have shown that butyrate can counteract obesity by increasing mitochondrial uncoupling and thereby promoting inefficient metabolism [197]. Butyrate is a promising candidate for the treatment of NDDs [198]. Indeed, different studies have shown that it has a neuroprotective effect in PD mice through indirect modulation of the gut microbiota, preventing inflammation in the gut–brain axis [199,200]. Moreover, by improving mitochondrial functions, butyrate counteracts the inflammatory processes and oxidative stress induced by an HFD in the cortex and synaptic area of the mouse brain. Thus, it may be involved in regulating neural plasticity and preventing the development of NDDs [200,201].

5.4. Endocannabinoid-like Compounds

In recent years, numerous researchers focused their attention on molecules with an endocannabinoid-like structure, known as endocannabinoid-like compounds, such as palmitoylethanolamide (PEA), oleoylethanolamide (OEA), and stearoyl ethanolamide (SEA), which have demonstrated both an anti-obesity effect and the ability to prevent the development of NDD. Despite numerous studies, the molecular mechanisms underlying these effects are still not fully understood. They are present in small quantities in food, but their main origin is endogenous synthesis [202].
PEA is an endogenous lipid mediator belonging to the N-acylethanolamine family, widely studied for its pleiotropic effects both centrally and peripherally. Several pre-clinical in vitro and in vivo studies indicate that PEA, an element naturally contained in some foods, can counteract obesity and be a potent agent for effective therapy of NDDs [203]. PEA reduced food intake, body weight, and body fat mass, effects mediated in part by restoring central and peripheral sensitivity to leptin in rats [204]. It has been observed that PEA stimulates the beige-conversion of the subcutaneous white adipose tissue (WAT) by increasing thermogenic markers and by PPAR-α activation. In addition, PEA can improve mitochondrial bioenergetics in mature adipocytes via AMPK phosphorylation [205]. It has been suggested that PEA could be a potential therapeutic agent for obesity-related neuropsychiatric comorbidities by regulating neuroinflammation, BBB breakdown, and neurotransmitter imbalance related to behavioral dysfunction [206]. Due to its antioxidant capabilities, PEA has been seen to enhance mitochondrial function in hippocampal regions and other tissues [207,208]. It also exerts its beneficial effects on neuroinflammation by acting on the composition of the gut microbiota, involving the microbiota–gut–brain axis [207].
OEA, an endocannabinoid-like compound, has been shown to have protective effects in many metabolic disorders by modulating the expression of genes involved in feeding behavior and lipid metabolism regulation [209]. Indeed, in rodents, intraperitoneal administration of OEA induces satiety, weight loss, stimulation of lipolysis, and enhancement of fatty acid oxidation by increasing the gene expression of proliferator-activated receptor-α (PPAR-α), a major player in the regulation of intra- and extracellular lipid metabolism [202,209]. Therefore, it is suggested as a potential therapeutic agent for the treatment of obesity. In addition, accumulating evidence has shown the protective role of OEA in inflammatory processes and NDDs. Indeed, it is an anti-inflammatory agent able to attenuate Aβ pathology in a mouse model of AD by modulating lipid metabolism and microglial phagocytosis [210]. OEA-mediated neuroprotection has been tested on in vivo and in vitro models of 6-hydroxydopamine (6-OH-DA)-induced degeneration, which causes Parkinsonian symptoms. In this animal model, administration of OEA prevents or alleviates Parkinsonian symptoms [211]. It was also tested against mitochondrial toxicity due to the electron transport chain complex II inhibitor, 3-nitropropionic acid (3-NP), in rat cortices, showing its ability to preserve mitochondrial functional integrity and cell viability [212]. It is therefore considered a potential agent for the treatment of NDDs.
SEA is a cannabinoid-like compound with a wide range of biological activities. It has anti-inflammatory properties that have been demonstrated in various animal models of pathological conditions. However, the molecular mechanisms are still unclear. Several studies have demonstrated the anorectic capacity of SEA in the mouse animal model. In addition, this anorectic effect is accompanied by a reduction in the mRNA expression of hepatic stearoyl-CoA desaturase-1 (SCD-1), which has recently been proposed as a molecular target against obesity. This suggests that SEA could have important implications in the pharmacological treatment of obesity [213]. SEA administration also had a positive impact on restoring the FFA composition of adipocytes taken from rats of different ages with HFD-induced insulin resistance, demonstrating its potential as a treatment for obesity-related complications [214]. In addition, SEA showed a neuroprotective effect against LPS-induced neuroinflammation in male C57BL/6 mice. In particular, by supporting blood–brain barrier integrity, SEA limited the spread of peripheral inflammation to the brain, thereby preventing the activation of resident microglia and the trafficking of leukocytes towards the brain parenchyma. Therefore, SEA can also be considered as a candidate for preventive therapy of cognitive dysfunction caused by neuroinflammation [215].

6. Conclusions

In recent years, a wealth of data has highlighted the correlation between obesity and NDDs. Although the exact underlying mechanisms remain unclear, it has been hypothesized that the adipose tissue dysfunction that occurs in obesity leads to systemic inflammation and to the alteration of the BBB resulting in neuroinflammation and cognitive decline. In this scenario, mitochondrial dysfunction plays a crucial role. Therefore, bioactive compounds that enhance mitochondrial function and eliminate excess ROS can be used to treat NDDs.
This review has discussed the beneficial effects of some bioactive food components in restoring and improving CNS mitochondrial function, supporting the importance of a healthy diet in preventing or slowing the onset and progression of NDDs. Among the bioactive compounds mentioned, polyphenols, CTs, and omega-3 PUFAs play an important role due to their antioxidant and anti-inflammatory effects, which can prevent ROS damage and improve mitochondrial function.
However, further studies are needed to define the exact molecular pathways underlying the onset and progression of NDDs and to elucidate the mechanisms driving the beneficial effects exerted by bioactive molecules in the treatment of mitochondrial dysfunction in NDDs. The results of these studies will allow setting up novel pharmacological approaches aimed at preventing the devastating progression of the aforementioned diseases. Therefore, this review encourages researchers to focus their attention on mitochondrial dysfunction in the development not only of NDDs but of all obesity-related pathological conditions.

Author Contributions

Conceptualization, G.C., F.M. and L.P.; writing—original draft preparation, G.C.; writing—review and editing, G.C., F.M., L.P., F.C., A.P., A.M., S.A., R.R., M.D., N.M., P.I., A.C., C.B. and B.P.; supervision, G.C., F.M., L.P. and F.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

Not applicable.

Acknowledgments

We thank BioRender (https://biorender.com) accessed on 23 and 24 December 2024, for providing the platform to create the scientific illustrations used in this work.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Boutari, C.; Mantzoros, C.S.A. Update on the Epidemiology of Obesity and a Call to Action: As Its Twin COVID-19 Pandemic Appears to Be Receding, the Obesity and Dysmetabolism Pandemic Continues to Rage On. Metabolism 2022, 133, 155217. [Google Scholar] [CrossRef] [PubMed]
  2. Yang, M.; Liu, S.; Zhang, C. The Related Metabolic Diseases and Treatments of Obesity. Healthcare 2022, 10, 1616. [Google Scholar] [CrossRef] [PubMed]
  3. Alsulami, S.; Baig, M.; Ahmad, T.; Althagafi, N.; Hazzazi, E.; Alsayed, R.; Alghamdi, M.; Almohammadi, T. Obesity Prevalence, Physical Activity, and Dietary Practices among Adults in Saudi Arabia. Front. Public Health 2023, 11, 1124051. [Google Scholar] [CrossRef]
  4. Neto, A.; Fernandes, A.; Barateiro, A. The Complex Relationship between Obesity and Neurodegenerative Diseases: An Updated Review. Front. Cell Neurosci. 2023, 17, 1294420. [Google Scholar] [CrossRef]
  5. Kershaw, E.E.; Flier, J.S. Adipose Tissue as an Endocrine Organ. J. Clin. Endocrinol. Metab. 2004, 89, 2548–2556. [Google Scholar] [CrossRef] [PubMed]
  6. Buckman, L.B.; Hasty, A.H.; Flaherty, D.K.; Buckman, C.T.; Thompson, M.M.; Matlock, B.K.; Weller, K.; Ellacott, K.L.J. Obesity Induced by a High-Fat Diet Is Associated with Increased Immune Cell Entry into the Central Nervous System. Brain Behav. Immun. 2014, 35, 33–42. [Google Scholar] [CrossRef]
  7. Heinonen, S.; Jokinen, R.; Rissanen, A.; Pietiläinen, K.H. White Adipose Tissue Mitochondrial Metabolism in Health and in Obesity. Obes. Rev. 2020, 21, e12958. [Google Scholar] [CrossRef]
  8. Lionetti, L.; Mollica, M.P.; Lombardi, A.; Cavaliere, G.; Gifuni, G.; Barletta, A. From Chronic Overnutrition to Insulin Resistance: The Role of Fat-Storing Capacity and Inflammation. Nutr. Metab. Cardiovasc. Dis. 2009, 19, 146–152. [Google Scholar] [CrossRef]
  9. Kirichenko, T.V.; Markina, Y.V.; Bogatyreva, A.I.; Tolstik, T.V.; Varaeva, Y.R.; Starodubova, A.V. The Role of Adipokines in Inflammatory Mechanisms of Obesity. Int. J. Mol. Sci. 2022, 23, 14982. [Google Scholar] [CrossRef]
  10. Mollica, M.P.; Lionetti, L.; Putti, R.; Cavaliere, G.; Gaita, M.; Barletta, A. From Chronic Overfeeding to Hepatic Injury: Role of Endoplasmic Reticulum Stress and Inflammation. Nutr. Metab. Cardiovasc. Dis. 2011, 21, 222–230. [Google Scholar] [CrossRef]
  11. Rogero, M.; Calder, P.O. Inflammation, Toll-Like Receptor 4 and Fatty Acids. Nutrients 2018, 10, 432. [Google Scholar] [CrossRef] [PubMed]
  12. Khanna, D.; Khanna, S.; Khanna, P.; Kahar, P.; Patel, B.M. Obesity: A Chronic Low-Grade Inflammation and Its Markers. Cureus 2022, 14, e22711. [Google Scholar] [CrossRef]
  13. Gómez-Apo, E.; Mondragón-Maya, A.; Ferrari-Díaz, M.; Silva-Pereyra, J. Structural Brain Changes Associated with Overweight and Obesity. J. Obes. 2021, 2021, 6613385. [Google Scholar] [CrossRef]
  14. ‘Ain Arshad, N.; Lin, T.S.; Yahaya, M.F. Metabolic Syndrome and Its Effect on the Brain: Possible Mechanism. CNSNDDT 2018, 17, 595–603. [Google Scholar] [CrossRef] [PubMed]
  15. Uranga, R.M.; Keller, J.N. The Complex Interactions Between Obesity, Metabolism and the Brain. Front. Neurosci. 2019, 13, 513. [Google Scholar] [CrossRef]
  16. Zhao, Y.; Gan, L.; Ren, L.; Lin, Y.; Ma, C.; Lin, X. Factors Influencing the Blood-Brain Barrier Permeability. Brain Res. 2022, 1788, 147937. [Google Scholar] [CrossRef] [PubMed]
  17. Kacířová, M.; Zmeškalová, A.; Kořínková, L.; Železná, B.; Kuneš, J.; Maletínská, L. Inflammation: Major Denominator of Obesity, Type 2 Diabetes and Alzheimer’s Disease-like Pathology? Clin. Sci. 2020, 134, 547–570. [Google Scholar] [CrossRef]
  18. Simi, A.; Tsakiri, N.; Wang, P.; Rothwell, N.J. Interleukin-1 and inflammatory neurodegeneration. Biochem. Soc. Trans. 2007, 35, 1122–1126. [Google Scholar] [CrossRef]
  19. Valdearcos, M.; Robblee, M.M.; Benjamin, D.I.; Nomura, D.K.; Xu, A.W.; Koliwad, S.K. Microglia Dictate the Impact of Saturated Fat Consumption on Hypothalamic Inflammation and Neuronal Function. Cell Rep. 2014, 6, 2124–2138. [Google Scholar] [CrossRef]
  20. Wang, Z.; Liu, D.; Wang, F.; Liu, S.; Zhao, S.; Ling, E.A.; Hao, A. Saturated Fatty Acids Activate Microglia via Toll-like Receptor 4/NF-κB Signalling. Br. J. Nutr. 2012, 2, 229–241. [Google Scholar] [CrossRef]
  21. Salsinha, A.S.; Socodato, R.; Relvas, J.B.; Pintado, M. The Pro- and Antiinflammatory Activity of Fatty Acids. In Bioactive Lipids; Elsevier: Amsterdam, The Netherlands, 2023; pp. 51–75. [Google Scholar]
  22. Kanoski, S.E.; Davidson, T.L. Western Diet Consumption and Cognitive Impairment: Links to Hippocampal Dysfunction and Obesity. Physiol. Behav. 2011, 103, 59–68. [Google Scholar] [CrossRef]
  23. Rasmussen Eid, H.; Rosness, T.A.; Bosnes, O.; Salvesen, Ø.; Knutli, M.; Stordal, E. Smoking and Obesity as Risk Factors in Frontotemporal Dementia and Alzheimer’s Disease: The HUNT Study. Dement. Geriatr. Cogn. Dis. Extra 2019, 9, 1–10. [Google Scholar] [CrossRef] [PubMed]
  24. Batarseh, N.; Al Thaher, Y. High-Fat Diet and Related Obesity Provoke Neurotoxins and Alter Neuro-Biomarkers Involved in Parkinson’s Disease. Obes. Med. 2023, 41, 100500. [Google Scholar] [CrossRef]
  25. Novo, A.M.; Batista, S. Multiple Sclerosis: Implications of Obesity in Neuroinflammation. In Obesity and Brain Function. Advances in Neurobiology; Springer: Cham, Switzerland, 2017; pp. 191–210. [Google Scholar]
  26. Anstey, K.J.; Cherbuin, N.; Budge, M.; Young, J. Body Mass Index in Midlife and Late-Life as a Risk Factor for Dementia: A Meta-Analysis of Prospective Studies. Obes. Rev. 2011, 12, e426–e437. [Google Scholar] [CrossRef] [PubMed]
  27. Hu, G.; Jousilahti, P.; Nissinen, A.; Antikainen, R.; Kivipelto, M.; Tuomilehto, J. Body Mass Index and the Risk of Parkinson Disease. Neurology 2006, 67, 1955–1959. [Google Scholar] [CrossRef]
  28. Gianfrancesco, M.A.; Barcellos, L.F. Obesity and Multiple Sclerosis Susceptibility: A Review. J. Neurol. Neuromed. 2016, 1, 1–5. [Google Scholar] [CrossRef] [PubMed]
  29. Mrak, R.E. Alzheimer-Type Neuropathological Changes in Morbidly Obese Elderly Individuals. Clin. Neuropathol. 2009, 28, 40–45. [Google Scholar] [CrossRef]
  30. Whitmer, R.A.; Gustafson, D.R.; Barrett-Connor, E.; Haan, M.N.; Gunderson, E.P.; Yaffe, K. Central Obesity and Increased Risk of Dementia More than Three Decades Later. Neurology 2008, 71, 1057–1064. [Google Scholar] [CrossRef]
  31. Nichols, E.; Szoeke, C.E.I.; Vollset, S.E.; Abbasi, N.; Abd-Allah, F.; Abdela, J.; Aichour, M.T.E.; Akinyemi, R.O.; Alahdab, F.; Asgedom, S.W.; et al. Global, Regional, and National Burden of Alzheimer’s Disease and Other Dementias, 1990–2016: A Systematic Analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019, 18, 88–106. [Google Scholar] [CrossRef]
  32. Hollands, C.; Bartolotti, N.; Lazarov, O. Alzheimer’s Disease and Hippocampal Adult Neurogenesis; Exploring Shared Mechanisms. Front. Neurosci. 2016, 10, 178. [Google Scholar] [CrossRef]
  33. Al-Kuraishy, H.M.; Al-Gareeb, A.I.; Alsayegh, A.A.; Hakami, Z.H.; Khamjan, N.A.; Saad, H.M.; Batiha, G.E.-S.; Waard, M. A Potential Link Between Visceral Obesity and Risk of Alzheimer’s Disease. Neurochem. Res. 2023, 48, 745–766. [Google Scholar] [CrossRef]
  34. Wan, Z.; Mah, D.; Simtchouk, S.; Kluftinger, A.; Little, J.P. Role of Amyloid β in the Induction of Lipolysis and Secretion of Adipokines from Human Adipose Tissue. Adipocyte 2015, 4, 212–216. [Google Scholar] [CrossRef] [PubMed]
  35. Mouton, P.R.; Chachich, M.E.; Quigley, C.; Spangler, E.; Ingram, D.K. Caloric Restriction Attenuates Amyloid Deposition in Middle-Aged Dtg APP/PS1 Mice. Neurosci. Lett. 2009, 464, 184–187. [Google Scholar] [CrossRef] [PubMed]
  36. Oliveira, T.P.D.; Morais, A.L.B.; Reis, P.L.B.; Palotás, A.; Vieira, L.B. A Potential Role for the Ketogenic Diet in Alzheimer’s Disease Treatment: Exploring Pre-Clinical and Clinical Evidence. Metabolites 2023, 14, 25. [Google Scholar] [CrossRef] [PubMed]
  37. Tabassum, S.; Misrani, A.; Yang, L. Exploiting Common Aspects of Obesity and Alzheimer’s Disease. Front. Hum. Neurosci. 2020, 14, 602360. [Google Scholar] [CrossRef]
  38. Julien, C.; Tremblay, C.; Phivilay, A.; Berthiaume, L.; Émond, V.; Julien, P.; Calon, F. High-Fat Diet Aggravates Amyloid-Beta and Tau Pathologies in the 3xTg-AD Mouse Model. Neurobiol. Aging 2010, 31, 1516–1531. [Google Scholar] [CrossRef]
  39. Lin, B.; Hasegawa, Y.; Takane, K.; Koibuchi, N.; Cao, C.; Kim-Mitsuyama, S. High-Fat-Diet Intake Enhances Cerebral Amyloid Angiopathy and Cognitive Impairment in a Mouse Model of Alzheimer’s Disease, Independently of Metabolic Disorders. J. Am. Heart Assoc. 2016, 5, e003154. [Google Scholar] [CrossRef]
  40. Maiti, P.; Manna, J.; Dunbar, G.L. Current Understanding of the Molecular Mechanisms in Parkinson’s Disease: Targets for Potential Treatments. Transl. Neurodegener. 2017, 6, 28. [Google Scholar] [CrossRef]
  41. Arenas, E. Parkinson’s Disease in the Single-Cell Era. Nat. Neurosci. 2022, 25, 536–538. [Google Scholar] [CrossRef]
  42. Hu, Q.; Wang, G. Mitochondrial Dysfunction in Parkinson’s Disease. Transl. Neurodegener. 2016, 5, 14. [Google Scholar] [CrossRef]
  43. Qu, Y.; Chen, X.; Xu, M.-M.; Sun, Q. Relationship between High Dietary Fat Intake and Parkinson’s Disease Risk: A Meta-Analysis. Neural Regen. Res. 2019, 14, 2156. [Google Scholar] [CrossRef]
  44. Tansey, M.G.; Wallings, R.L.; Houser, M.C.; Herrick, M.K.; Keating, C.E.; Joers, V. Inflammation and Immune Dysfunction in Parkinson Disease. Nat. Rev. Immunol. 2022, 22, 657–673. [Google Scholar] [CrossRef] [PubMed]
  45. Doria, M.; Maugest, L.; Moreau, T.; Lizard, G.; Vejux, A. Contribution of Cholesterol and Oxysterols to the Pathophysiology of Parkinson’s Disease. Free Radic. Biol. Med. 2016, 101, 393–400. [Google Scholar] [CrossRef]
  46. Han, J.; Nepal, P.; Odelade, A.; Freely, F.D.; Belton, D.M.; Graves, J.L.; Maldonado-Devincci, A.M. High-Fat Diet-Induced Weight Gain, Behavioral Deficits, and Dopamine Changes in Young C57BL/6J Mice. Front. Nutr. 2021, 7, 591161. [Google Scholar] [CrossRef] [PubMed]
  47. Narayanaswami, V.; Thompson, A.C.; Cassis, L.A.; Bardo, M.T.; Dwoskin, L.P. Diet-Induced Obesity: Dopamine Transporter Function, Impulsivity and Motivation. Int. J. Obes. 2013, 37, 1095–1103. [Google Scholar] [CrossRef]
  48. Barry, R.L.; Byun, N.E.; Williams, J.M.; Siuta, M.A.; Tantawy, M.N.; Speed, N.K.; Saunders, C.; Galli, A.; Niswender, K.D.; Avison, M.J. Brief Exposure to Obesogenic Diet Disrupts Brain Dopamine Networks. PLoS ONE 2018, 13, e0191299. [Google Scholar] [CrossRef] [PubMed]
  49. Correia, I.; Cunha, C.; Bernardes, C.; Nunes, C.; Macário, C.; Sousa, L.; Batista, S. Prevalence, Incidence, and Mortality of Multiple Sclerosis in Coimbra, Portugal. Neuroepidemiology 2024, 58, 57–63. [Google Scholar] [CrossRef]
  50. Oh, J.; Vidal-Jordana, A.; Montalban, X. Multiple Sclerosis: Clinical Aspects. Curr. Opin. Neurol. 2018, 31, 752–759. [Google Scholar] [CrossRef]
  51. Munger, K.L. Childhood Obesity Is a Risk Factor for Multiple Sclerosis. Mult. Scler. J. 2013, 19, 1800. [Google Scholar] [CrossRef]
  52. Marrie, R.; Horwitz, R.; Cutter, G.; Tyry, T.; Campagnolo, D.; Vollmer, T. High Frequency of Adverse Health Behaviors in Multiple Sclerosis. Mult. Scler. J. 2009, 15, 105–113. [Google Scholar] [CrossRef]
  53. Marrie, R.A.; Horwitz, R.I.; Cutter, G.; Tyry, T.; Vollmer, T. Association between Comorbidity and Clinical Characteristics of MS. Acta Neurol. Scand. 2011, 124, 135–141. [Google Scholar] [CrossRef]
  54. Palavra, F.; Marado, D.; Mascarenhas-Melo, F.; Sereno, J.; Teixeira-Lemos, E.; Nunes, C.C.; Gonçalves, G.; Teixeira, F.; Reis, F. New Markers of Early Cardiovascular Risk in Multiple Sclerosis Patients: Oxidized-LDL Correlates with Clinical Staging. Dis. Markers 2013, 34, 341–348. [Google Scholar] [CrossRef]
  55. Matarese, G.; Carrieri, P.B.; Montella, S.; Rosa, V.; Cava, A. Leptin as a Metabolic Link to Multiple Sclerosis. Nat. Rev. Neurol. 2010, 6, 455–461. [Google Scholar] [CrossRef]
  56. Emamgholipour, S.; Eshaghi, S.M.; Hossein-nezhad, A.; Mirzaei, K.; Maghbooli, Z.; Sahraian, M.A. Adipocytokine Profile, Cytokine Levels and Foxp3 Expression in Multiple Sclerosis: A Possible Link to Susceptibility and Clinical Course of Disease. PLoS ONE 2013, 8, e76555. [Google Scholar] [CrossRef]
  57. Piccio, L.; Stark, J.L.; Cross, A.H. Chronic Calorie Restriction Attenuates Experimental Autoimmune Encephalomyelitis. J. Leukoc. Biol. 2008, 84, 940–948. [Google Scholar] [CrossRef]
  58. Lee, K.H.; Cha, M.; Lee, B.H. Neuroprotective Effect of Antioxidants in the Brain. Int. J. Mol. Sci. 2020, 21, 7152. [Google Scholar] [CrossRef]
  59. Nissanka, N.; Moraes, C.T. Mitochondrial DNA Damage and Reactive Oxygen Species in Neurodegenerative Disease. FEBS Lett. 2018, 5, 728–742. [Google Scholar] [CrossRef]
  60. Schöttl, T.; Kappler, L.; Fromme, T.; Klingenspor, M. Limited OXPHOS Capacity in White Adipocytes Is a Hallmark of Obesity in Laboratory Mice Irrespective of the Glucose Tolerance Status. Mol. Metab. 2015, 4, 631–642. [Google Scholar] [CrossRef]
  61. Cavaliere, G.; Cimmino, F.; Trinchese, G.; Catapano, A.; Petrella, L.; D’Angelo, M.; Lucchin, L.; Mollica, M.P. From Obesity-Induced Low-Grade Inflammation to Lipotoxicity and Mitochondrial Dysfunction: Altered Multi-Crosstalk between Adipose Tissue and Metabolically Active Organs. Antioxidants 2023, 12, 1172. [Google Scholar] [CrossRef]
  62. Nadler, S.T.; Stoehr, J.P.; Schueler, K.L.; Tanimoto, G.; Yandell, B.S.; Attie, A.D. The Expression of Adipogenic Genes Is Decreased in Obesity and Diabetes Mellitus. Proc. Natl. Acad. Sci. USA 2000, 97, 11371–11376. [Google Scholar] [CrossRef]
  63. Todosenko, N.; Khaziakhmatova, O.; Malashchenko, V.; Yurova, K.; Bograya, M.; Beletskaya, M.; Vulf, M.; Gazatova, N.; Litvinova, L. Mitochondrial Dysfunction Associated with MtDNA in Metabolic Syndrome and Obesity. Int. J. Mol. Sci. 2023, 24, 12012. [Google Scholar] [CrossRef]
  64. Fischer, B.; Schöttl, T.; Schempp, C.; Fromme, T.; Hauner, H.; Klingenspor, M.; Skurk, T. Inverse Relationship between Body Mass Index and Mitochondrial Oxidative Phosphorylation Capacity in Human Subcutaneous Adipocytes. Am. J. Physiol.-Endocrinol. Metab. 2015, 309, 380–387. [Google Scholar] [CrossRef]
  65. Gao, C.-L.; Zhu, C.; Zhao, Y.-P.; Chen, X.-H.; Ji, C.-B.; Zhang, C.-M.; Zhu, J.-G.; Xia, Z.-K.; Tong, M.-L.; Guo, X.-R. Mitochondrial Dysfunction Is Induced by High Levels of Glucose and Free Fatty Acids in 3T3-L1 Adipocytes. Mol. Cell. Endocrinol. 2010, 320, 25–33. [Google Scholar] [CrossRef] [PubMed]
  66. Hernández-Aguilera, A.; Rull, A.; Rodríguez-Gallego, E.; Riera-Borrull, M.; Luciano-Mateo, F.; Camps, J.; Menéndez, J.A.; Joven, J. Mitochondrial Dysfunction: A Basic Mechanism in Inflammation-Related Non-Communicable Diseases and Therapeutic Opportunities. Mediat. Inflamm. 2013, 2013, 135698. [Google Scholar] [CrossRef]
  67. Cinti, S.; Mitchell, G.; Barbatelli, G.; Murano, I.; Ceresi, E.; Faloia, E.; Wang, S.; Fortier, M.; Greenberg, A.S.; Obin, M.S. Adipocyte Death Defines Macrophage Localization and Function in Adipose Tissue of Obese Mice and Humans. J. Lipid Res. 2005, 46, 2347–2355. [Google Scholar] [CrossRef] [PubMed]
  68. Trinchese, G.; Cimmino, F.; Catapano, A.; Cavaliere, G.; Mollica, M.P. Mitochondria: The Gatekeepers between Metabolism and Immunity. Front. Immunol. 2024, 15, 1334006. [Google Scholar] [CrossRef] [PubMed]
  69. Woo, C.-Y.; Jang, J.E.; Lee, S.E.; Koh, E.H.; Lee, K.-U. Mitochondrial Dysfunction in Adipocytes as a Primary Cause of Adipose Tissue Inflammation. Diabetes Metab. J. 2019, 43, 247. [Google Scholar] [CrossRef]
  70. Cavaliere, G.; Catapano, A.; Trinchese, G.; Cimmino, F.; Menale, C.; Petrella, L.; Mollica, M.P. Crosstalk between Adipose Tissue and Hepatic Mitochondria in the Development of the Inflammation and Liver Injury during Ageing in High-Fat Diet Fed Rats. Int. J. Mol. Sci. 2023, 24, 2967. [Google Scholar] [CrossRef]
  71. Mollica, M.P.; Lionetti, L.; Crescenzo, R.; D’Andrea, E.; Ferraro, M.; Liverini, G.; Iossa, S. Heterogeneous Bioenergetic Behaviour of Subsarcolemmal and Intermyofibrillar Mitochondria in Fed and Fasted Rats. Cell. Mol. Life Sci. 2006, 63, 358–366. [Google Scholar] [CrossRef]
  72. Erecinska, M.; Cherian, S.; Silver, I.A. Energy Metabolism in Mammalian Brain during Development. Prog. Neurobiol. 2004, 73, 397–445. [Google Scholar] [CrossRef]
  73. Misgeld, T.; Schwarz, T.L. Mitostasis in Neurons: Maintaining Mitochondria in an Extended Cellular Architecture. Neuron 2017, 96, 651–666. [Google Scholar] [CrossRef]
  74. Attwell, D.; Laughlin, S.B. An Energy Budget for Signaling in the Grey Matter of the Brain. J. Cereb. Blood Flow. Metab. 2001, 21, 1133–1145. [Google Scholar] [CrossRef] [PubMed]
  75. Perrone-Capano, C.; Volpicelli, F.; Penna, E.; Chun, J.T.; Crispino, M. Presynaptic Protein Synthesis and Brain Plasticity: From Physiology to Neuropathology. Prog. Neurobiol. 2021, 202, 102051. [Google Scholar] [CrossRef]
  76. Penna, E.; Cerciello, A.; Chambery, A.; Russo, R.; Cernilogar, F.M.; Pedone, E.M.; Perrone-Capano, C.; Cappello, S.; Giaimo, R.; Crispino, M. Cystatin B Involvement in Synapse Physiology of Rodent Brains and Human Cerebral Organoids. Front. Mol. Neurosci. 2019, 12, 195. [Google Scholar] [CrossRef]
  77. Citri, A.; Malenka, R.C. Synaptic Plasticity: Multiple Forms, Functions, and Mechanisms. Neuropsychopharmacology 2008, 33, 18–41. [Google Scholar] [CrossRef] [PubMed]
  78. Alnaes, E.; Rahamimoff, R. On the Role of Mitochondria in Transmitter Release from Motor Nerve Terminals. J. Physiol. 1975, 248, 285–306. [Google Scholar] [CrossRef] [PubMed]
  79. Cherra, S.J.; Steer, E.; Gusdon, A.M.; Kiselyov, K.; Chu, C.T. Mutant LRRK2 Elicits Calcium Imbalance and Depletion of Dendritic Mitochondria in Neurons. Am. J. Pathol. 2013, 182, 474–484. [Google Scholar] [CrossRef]
  80. Cavaliere, G.; Trinchese, G.; Penna, E.; Cimmino, F.; Pirozzi, C.; Lama, A.; Annunziata, C.; Catapano, A.; Mattace Raso, G.; Meli, R.; et al. High-Fat Diet Induces Neuroinflammation and Mitochondrial Impairment in Mice Cerebral Cortex and Synaptic Fraction. Front. Cell. Neurosci. 2019, 13, 509. [Google Scholar] [CrossRef]
  81. Penna, E.; Pizzella, A.; Cimmino, F.; Trinchese, G.; Cavaliere, G.; Catapano, A.; Allocca, I.; Chun, J.T.; Campanozzi, A.; Messina, G. Neurodevelopmental Disorders: Effect of High-Fat Diet on Synaptic Plasticity and Mitochondrial Functions. Brain Sci. 2020, 10, 805. [Google Scholar] [CrossRef]
  82. Bliss, T.V.P.; Collingridge, G.L.; Morris, R.G.M. Synaptic Plasticity in Health and Disease: Introduction and Overview. Philos. Trans. R. Soc. B Biol. Sci. 2014, 369, 20130129. [Google Scholar] [CrossRef]
  83. Trinchese, G.; Cimmino, F.; Cavaliere, G.; Catapano, A.; Fogliano, C.; Lama, A.; Pirozzi, C.; Cristiano, C.; Russo, R.; Petrella, L. The Hepatic Mitochondrial Alterations Exacerbate Meta-Inflammation in Autism Spectrum Disorders. Antioxidants 2022, 11, 1990. [Google Scholar] [CrossRef] [PubMed]
  84. Matteo, F.; Pipicelli, F.; Kyrousi, C.; Tovecci, I.; Penna, E.; Crispino, M.; Chambery, A.; Russo, R.; Ayo-Martin, A.C.; Giordano, M. Cystatin B Is Essential for Proliferation and Interneuron Migration in Individuals with EPM 1 Epilepsy. EMBO Mol. Med. 2020, 12, e11419. [Google Scholar] [CrossRef]
  85. Sanz-Alcázar, A.; Britti, E.; Delaspre, F.; Medina-Carbonero, M.; Pazos-Gil, M.; Tamarit, J.; Ros, J.; Cabiscol, E. Mitochondrial Impairment, Decreased Sirtuin Activity and Protein Acetylation in Dorsal Root Ganglia in Friedreich Ataxia Models. Cell. Mol. Life Sci. 2024, 81, 12. [Google Scholar] [CrossRef] [PubMed]
  86. Ma, W.; Yuan, L.; Yu, H.; Xi, Y.; Xiao, R. Mitochondrial Dysfunction and Oxidative Damage in the Brain of Diet-Induced Obese Rats but Not in Diet-Resistant Rats. Life Sci. 2014, 110, 53–60. [Google Scholar] [CrossRef] [PubMed]
  87. Korshunov, S.S.; Skulachev, V.P.; Starkov, A.A. High protonic potential actuates a mechanism of production of reactive oxygen species in mitochondria. FEBS Lett. 1997, 416, 15–18. [Google Scholar] [CrossRef]
  88. Cavaliere, G.; Viggiano, E.; Trinchese, G.; De Filippo, C.; Messina, A.; Monda, V.; Valenzano, A.; Cincione, R.I.; Zammit, C.; Cimmino, F.; et al. Long Feeding High-Fat Diet Induces Hypothalamic Oxidative Stress and Inflammation, and Prolonged Hypothalamic AMPK Activation in Rat Animal Model. Front. Physiol. 2018, 9, 818. [Google Scholar] [CrossRef]
  89. Park, H.-S.; Cho, H.-S.; Kim, T.-W. Physical Exercise Promotes Memory Capability by Enhancing Hippocampal Mitochondrial Functions and Inhibiting Apoptosis in Obesity-Induced Insulin Resistance by High Fat Diet. Metab. Brain Dis. 2018, 33, 283–292. [Google Scholar] [CrossRef]
  90. Siino, V.; Jensen, P.; James, P.; Vasto, S.; Amato, A.; Mulè, F.; Accardi, G.; Larsen, M.R. Obesogenic Diets Cause Alterations on Proteins and Theirs Post-Translational Modifications in Mouse Brains. Nutr. Metab. Insights 2021, 14, 117863882110124. [Google Scholar] [CrossRef]
  91. Zhang, L.; Wu, J.; Zhu, Z.; He, Y.; Fang, R. Mitochondrion: A Bridge Linking Aging and Degenerative Diseases. Life Sci. 2023, 322, 121666. [Google Scholar] [CrossRef]
  92. Gowda, P.; Reddy, P.H.; Kumar, S. Deregulated Mitochondrial MicroRNAs in Alzheimer’s Disease: Focus on Synapse and Mitochondria. Ageing Res. Rev. 2022, 73, 101529. [Google Scholar] [CrossRef]
  93. Gibson, G.E.; Sheu, K.-F.R.; Blass, J.P.; Baker, A.; Carlson, K.C.; Harding, B.; Perrino, P. Reduced Activities of Thiamine-Dependent Enzymes in the Brains and Peripheral Tissues of Patients With Alzheimer’s Disease. Arch. Neurol. 1988, 45, 836–840. [Google Scholar] [CrossRef] [PubMed]
  94. Casley, C.S.; Canevari, L.; Land, J.M.; Clark, J.B.; Sharpe, M.A. Β-Amyloid Inhibits Integrated Mitochondrial Respiration and Key Enzyme Activities. J. Neurochem. 2002, 80, 91–100. [Google Scholar] [CrossRef]
  95. Gonzalez-Garcia, M.; Fusco, G.; Simone, A. Membrane Interactions and Toxicity by Misfolded Protein Oligomers. Front. Cell Dev. Biol. 2021, 9, 642623. [Google Scholar] [CrossRef] [PubMed]
  96. Leal, N.S.; Dentoni, G.; Schreiner, B.; Naia, L.; Piras, A.; Graff, C.; Cattaneo, A.; Meli, G.; Hamasaki, M.; Nilsson, P. Amyloid β-Peptide Increases Mitochondria-Endoplasmic Reticulum Contact Altering Mitochondrial Function and Autophagosome Formation in Alzheimer’s Disease-Related Models. Cells 2020, 9, 2552. [Google Scholar] [CrossRef]
  97. Hu, W.; Wang, Z.; Zheng, H. Mitochondrial Accumulation of Amyloid β (Aβ) Peptides Requires TOMM22 as a Main Aβ Receptor in Yeast. J. Biol. Chem. 2018, 293, 12681–12689. [Google Scholar] [CrossRef]
  98. Koss, D.J.; Jones, G.; Cranston, A.; Gardner, H.; Kanaan, N.M.; Platt, B. Soluble Pre-Fibrillar Tau and β-Amyloid Species Emerge in Early Human Alzheimer’s Disease and Track Disease Progression and Cognitive Decline. Acta Neuropathol. 2016, 132, 875–895. [Google Scholar] [CrossRef]
  99. Pérez, M.J.; Jara, C.; Quintanilla, R.A. Contribution of Tau Pathology to Mitochondrial Impairment in Neurodegeneration. Front. Neurosci. 2018, 12, 441. [Google Scholar] [CrossRef] [PubMed]
  100. Cieri, D.; Vicario, M.; Vallese, F.; D’Orsi, B.; Berto, P.; Grinzato, A.; Catoni, C.; Stefani, D.; Rizzuto, R.; Brini, M. Tau Localises within Mitochondrial Sub-Compartments and Its Caspase Cleavage Affects ER-Mitochondria Interactions and Cellular Ca2+ Handling. Biochim. Et Biophys. Acta (BBA)—Mol. Basis Dis. 2018, 1864, 3247–3256. [Google Scholar] [CrossRef]
  101. Salvadores, N.; Gerónimo-Olvera, C.; Court, F.A. Axonal Degeneration in AD: The Contribution of Aβ and Tau. Front. Aging Neurosci. 2020, 12, 581767. [Google Scholar] [CrossRef]
  102. Kopeikina, K.J.; Carlson, G.A.; Pitstick, R.; Ludvigson, A.E.; Peters, A.; Luebke, J.I.; Koffie, R.M.; Frosch, M.P.; Hyman, B.T.; Spires-Jones, T.L. Tau Accumulation Causes Mitochondrial Distribution Deficits in Neurons in a Mouse Model of Tauopathy and in Human Alzheimer’s Disease Brain. Am. J. Pathol. 2011, 179, 2071–2082. [Google Scholar] [CrossRef]
  103. Wang, X.; Su, B.; Lee, H.-G.; Li, X.; Perry, G.; Smith, M.A.; Zhu, X. Impaired Balance of Mitochondrial Fission and Fusion in Alzheimer’s Disease. J. Neurosci. 2009, 29, 9090–9103. [Google Scholar] [CrossRef] [PubMed]
  104. Manczak, M.; Reddy, P.H. Abnormal Interaction between the Mitochondrial Fission Protein Drp1 and Hyperphosphorylated Tau in Alzheimer’s Disease Neurons: Implications for Mitochondrial Dysfunction and Neuronal Damage. Hum. Mol. Genet. 2012, 21, 2538–2547. [Google Scholar] [CrossRef] [PubMed]
  105. Wang, X.; Su, B.; Siedlak, S.L.; Moreira, P.I.; Fujioka, H.; Wang, Y.; Casadesus, G.; Zhu, X. Amyloid-β Overproduction Causes Abnormal Mitochondrial Dynamics via Differential Modulation of Mitochondrial Fission/Fusion Proteins. Proc. Natl. Acad. Sci. USA 2008, 105, 19318–19323. [Google Scholar] [CrossRef] [PubMed]
  106. Grünewald, A.; Kumar, K.R.; Sue, C.M. New Insights into the Complex Role of Mitochondria in Parkinson’s Disease. Prog. Neurobiol. 2019, 177, 73–93. [Google Scholar] [CrossRef]
  107. Winklhofer, K.F.; Haass, C. Mitochondrial Dysfunction in Parkinson’s Disease. Biochim. Et Biophys. Acta (BBA)—Mol. Basis Disease 2010, 1802, 29–44. [Google Scholar] [CrossRef]
  108. Ludtmann, M.H.R.; Abramov, A.Y. Mitochondrial Calcium Imbalance in Parkinson’s Disease. Neurosci. Lett. 2018, 663, 86–90. [Google Scholar] [CrossRef]
  109. Tretter, L.; Sipos, I.; Adam-Vizi, V. Initiation of Neuronal Damage by Complex I Deficiency and Oxidative Stress in Parkinson’s Disease. Neurochem. Res. 2004, 29, 569–577. [Google Scholar] [CrossRef]
  110. Ortiz, G.G.; Pacheco-Moisés, F.P.; Gómez-Rodríguez, V.M.; González-Renovato, E.D.; Torres-Sánchez, E.D.; Ramírez-Anguiano, A.C. Fish Oil, Melatonin and Vitamin E Attenuates Midbrain Cyclooxygenase-2 Activity and Oxidative Stress after the Administration of 1-Methyl-4-Phenyl-1,2,3,6- Tetrahydropyridine. Metab. Brain Dis. 2013, 28, 705–709. [Google Scholar] [CrossRef]
  111. Ge, P.; Dawson, V.L.; Dawson, T.M. PINK1 and Parkin Mitochondrial Quality Control: A Source of Regional Vulnerability in Parkinson’s Disease. Mol. Neurodegener. 2020, 15, 20. [Google Scholar] [CrossRef]
  112. Deng, H.; Dodson, M.W.; Huang, H.; Guo, M. The Parkinson’s Disease Genes Pink1 and Parkin Promote Mitochondrial Fission and/or Inhibit Fusion in Drosophila. Proc. Natl. Acad. Sci. USA 2008, 105, 14503–14508. [Google Scholar] [CrossRef]
  113. Palacino, J.J.; Sagi, D.; Goldberg, M.S.; Krauss, S.; Motz, C.; Wacker, M.; Klose, J.; Shen, J. Mitochondrial Dysfunction and Oxidative Damage in Parkin-Deficient Mice. J. Biol. Chem. 2004, 279, 18614–18622. [Google Scholar] [CrossRef] [PubMed]
  114. Morais, V.A.; Verstreken, P.; Roethig, A.; Smet, J.; Snellinx, A.; Vanbrabant, M.; Haddad, D.; Frezza, C.; Mandemakers, W.; Vogt-Weisenhorn, D. Parkinson’s Disease Mutations in PINK1 Result in Decreased Complex I Activity and Deficient Synaptic Function. EMBO Mol. Med. 2009, 1, 99–111. [Google Scholar] [CrossRef]
  115. Filichia, E.; Hoffer, B.; Qi, X.; Luo, Y. Inhibition of Drp1 Mitochondrial Translocation Provides Neural Protection in Dopaminergic System in a Parkinson’s Disease Model Induced by MPTP. Sci. Rep. 2016, 6, 32656. [Google Scholar] [CrossRef] [PubMed]
  116. Bonifati, V.; Rizzu, P.; Squitieri, F.; Krieger, E.; Vanacore, N.; Swieten, J.C.; Brice, A.; Duijn, C.M.; Oostra, B.; Meco, G. DJ-1(PARK7), a Novel Gene for Autosomal Recessive, Early Onset Parkinsonism. Neurol. Sci. 2003, 24, 159–160. [Google Scholar] [CrossRef]
  117. Irrcher, I.; Aleyasin, H.; Seifert, E.L.; Hewitt, S.J.; Chhabra, S.; Phillips, M.; Lutz, A.K.; Rousseaux, M.W.C.; Bevilacqua, L.; Jahani-Asl, A. Loss of the Parkinson’s Disease-Linked Gene DJ-1 Perturbs Mitochondrial Dynamics. Hum. Mol. Genet. 2010, 19, 3734–3746. [Google Scholar] [CrossRef]
  118. Luth, E.S.; Stavrovskaya, I.G.; Bartels, T.; Kristal, B.S.; Selkoe, D.J.S. Prefibrillar α-Synuclein Oligomers Promote Complex I-Dependent, Ca2+-Induced Mitochondrial Dysfunction. J. Biol. Chem. 2014, 289, 21490–21507. [Google Scholar] [CrossRef]
  119. Subramaniam, S.R.; Vergnes, L.; Franich, N.R.; Reue, K.; Chesselet, M.-F. Region Specific Mitochondrial Impairment in Mice with Widespread Overexpression of Alpha-Synuclein. Neurobiol. Dis. 2014, 70, 204–213. [Google Scholar] [CrossRef] [PubMed]
  120. Chinta, S.J.; Mallajosyula, J.K.; Rane, A.; Andersen, J.K. Mitochondrial Alpha-Synuclein Accumulation Impairs Complex I Function in Dopaminergic Neurons and Results in Increased Mitophagy in Vivo. Neurosci. Lett. 2010, 486, 235–239. [Google Scholar] [CrossRef]
  121. Risiglione, P.; Zinghirino, F.; Di Rosa, M.C.; Magrì, A.; Messina, A. Alpha-Synuclein and Mitochondrial Dysfunction in Parkinson’s Disease: The Emerging Role of VDAC. Biomolecules 2021, 11, 718. [Google Scholar] [CrossRef]
  122. Ludtmann, M.H.R.; Angelova, P.R.; Ninkina, N.N.; Gandhi, S.; Buchman, V.L.; Abramov, A.Y. Monomeric Alpha-Synuclein Exerts a Physiological Role on Brain ATP Synthase. J. Neurosci. 2016, 36, 10510–10521. [Google Scholar] [CrossRef]
  123. Ludtmann, M.H.R.; Angelova, P.R.; Horrocks, M.H.; Choi, M.L.; Rodrigues, M.; Baev, A.Y.; Berezhnov, A.V.; Yao, Z.; Little, D.; Banushi, B. α-Synuclein Oligomers Interact with ATP Synthase and Open the Permeability Transition Pore in Parkinson’s Disease. Nat. Commun. 2018, 9, 2293. [Google Scholar] [CrossRef] [PubMed]
  124. Di Maio, R.; Barrett, P.J.; Hoffman, E.K.; Barrett, C.W.; Zharikov, A.; Borah, A.; Hu, X.; McCoy, J.; Chu, C.T.; Burton, E.A.; et al. α-Synuclein binds to TOM20 and inhibits mitochondrial protein import in Parkinson’s disease. Sci. Transl. Med. 2016, 8, 342ra78. [Google Scholar] [CrossRef] [PubMed]
  125. López-Muguruza, E.; Matute, C. Alterations of Oligodendrocyte and Myelin Energy Metabolism in Multiple Sclerosis. Int. J. Mol. Sci. 2023, 24, 12912. [Google Scholar] [CrossRef] [PubMed]
  126. Li, S.; Clements, R.; Sulak, M.; Gregory, R.; Freeman, E.; McDonough, J. Decreased NAA in gray matter is correlated with decreased availability of acetate in white matter in postmortem multiple sclerosis cortex. Neurochem. Res. 2013, 38, 2385–2396. [Google Scholar] [CrossRef]
  127. Licht-Mayer, S.; Campbell, G.R.; Canizares, M.; Mehta, A.R.; Gane, A.B.; McGill, K.; Ghosh, A.; Fullerton, A.; Menezes, N.; Dean, J. Enhanced Axonal Response of Mitochondria to Demyelination Offers Neuroprotection: Implications for Multiple Sclerosis. Acta Neuropathol. 2020, 140, 143–167. [Google Scholar] [CrossRef]
  128. Dutta, R.; McDonough, J.; Yin, X.; Peterson, J.; Chang, A.; Torres, T.; Gudz, T.; Macklin, W.B.; Lewis, D.A.; Fox, R.J. Mitochondrial Dysfunction as a Cause of Axonal Degeneration in Multiple Sclerosis Patients. Ann. Neurol. 2006, 59, 478–489. [Google Scholar] [CrossRef]
  129. Mancini, A.; Gaetani, L.; Gentili, L.; Filippo, M. Finding a Way to Preserve Mitochondria: New Pathogenic Pathways in Experimental Multiple Sclerosis. Neural Regen. Res. 2019, 14, 77. [Google Scholar] [CrossRef] [PubMed]
  130. Campbell, G.; Licht-Mayer, S.; Mahad, D. Targeting Mitochondria to Protect Axons in Progressive MS. Neurosci. Lett. 2019, 710, 134258. [Google Scholar] [CrossRef]
  131. Smith, K.J.; Kapoor, R.; Hall, S.M.; Davies, M. Electrically Active Axons Degenerate When Exposed to Nitric Oxide. Ann. Neurol. 2001, 49, 470–476. [Google Scholar] [CrossRef]
  132. Ghafourifar, P.; Mousavizadeh, K.; Parihar, M.S.; Nazarewicz, R.R.; Parihar, A.; Zenebe, W.J. Mitochondria in multiple sclerosis. Front. Biosci. 2008, 13, 3116–3126. [Google Scholar] [CrossRef]
  133. Witte, M.E.; Nijland, P.G.; Drexhage, J.A.; Gerritsen, W.; Geerts, D.; van Het Hof, B.; Reijerkerk, A.; de Vries, H.E.; van der Valk, P.; van Horssen, J. Reduced expression of PGC-1α partly underlies mitochondrial changes and correlates with neuronal loss in multiple sclerosis cortex. Acta Neuropathol. 2013, 125, 231–243. [Google Scholar] [CrossRef] [PubMed]
  134. Witte, M.E.; Bø, L.; Rodenburg, R.J.; Belien, J.A.; Musters, R.; Hazes, T.; Wintjes, L.T.; Smeitink, J.A.; Geurts, J.J.; De Vries, H.E.; et al. Enhanced number and activity of mitochondria in multiple sclerosis lesions. J. Pathol. 2009, 219, 193–204. [Google Scholar] [CrossRef] [PubMed]
  135. Castellazzi, M.; Patergnani, S.; Donadio, M.; Giorgi, C.; Bonora, M.; Fainardi, E.; Casetta, I.; Granieri, E.; Pugliatti, M.; Pinton, P. Correlation between auto/mitophagic processes and magnetic resonance imaging activity in multiple sclerosis patients. J. Neuroinflamm. 2019, 16, 131. [Google Scholar] [CrossRef]
  136. Luo, F.; Herrup, K.; Qi, X.; Yang, Y. Inhibition of Drp1 Hyper-Activation Is Protective in Animal Models of Experimental Multiple Sclerosis. Exp. Neurol. 2017, 292, 21–34. [Google Scholar] [CrossRef]
  137. Thai, T.Q.; Nguyen, H.B.; Sui, Y.; Ikenaka, K.; Oda, T.; Ohno, N. Interactions between Mitochondria and Endoplasmic Reticulum in Demyelinated Axons. Med. Mol. Morphol. 2019, 52, 135–146. [Google Scholar] [CrossRef]
  138. Lama, A.; Pirozzi, C.; Mollica, M.P.; Trinchese, G.; Guida, F.; Cavaliere, G.; Calignano, A.; Mattace Raso, G.; Berni Canani, R.; Meli, R. Polyphenol-Rich Virgin Olive Oil Reduces Insulin Resistance and Liver Inflammation and Improves Mitochondrial Dysfunction in High-Fat Diet Fed Rats. Mol. Nutr. Food Res. 2017, 61, 1600418. [Google Scholar] [CrossRef] [PubMed]
  139. Yan, L.; Guo, M.-S.; Zhang, Y.; Yu, L.; Wu, J.-M.; Tang, Y.; Ai, W.; Zhu, F.-D.; Law, B.Y.-K.; Chen, Q. Dietary Plant Polyphenols as the Potential Drugs in Neurodegenerative Diseases: Current Evidence. Advances, and Opportunities. Oxidative Med. Cell. Longev. 2022, 2022, 5288698. [Google Scholar] [CrossRef]
  140. De Oliveira, M.R.; Nabavi, S.F.; Manayi, A.; Daglia, M.; Hajheydari, Z.; Nabavi, S.M. Resveratrol and the Mitochondria: From Triggering the Intrinsic Apoptotic Pathway to Inducing Mitochondrial Biogenesis, a Mechanistic View. Biochim. Et Biophys. Acta (BBA)—General. Subjects 2016, 1860, 727–745. [Google Scholar] [CrossRef]
  141. Lasa, A.; Churruca, I.; Eseberri, I.; Andrés-Lacueva, C.; Portillo, M.P. Delipidating effect of resveratrol metabolites in 3T3-L1 adipocytes. Mol. Nutr. Food Res. 2012, 10, 1559–1568. [Google Scholar] [CrossRef]
  142. Rivera, L.; Morón, R.; Zarzuelo, A.; Galisteo, M. Long-Term Resveratrol Administration Reduces Metabolic Disturbances and Lowers Blood Pressure in Obese Zucker Rats. Biochem. Pharmacol. 2009, 6, 1053–1063. [Google Scholar] [CrossRef]
  143. Moseti, D.; Regassa, A.; Kim, W.K. Molecular Regulation of Adipogenesis and Potential Anti-Adipogenic Bioactive Molecules. Int. J. Mol. Sci. 2016, 1, 124. [Google Scholar] [CrossRef] [PubMed]
  144. Shaito, A.; Al-Mansoob, M.; Ahmad, S.M.S.; Haider, M.Z.; Eid, A.H.; Posadino, A.M.; Pintus, G.; Giordo, R. Resveratrol-Mediated Regulation of Mitochondria Biogenesis-Associated Pathways in Neurodegenerative Diseases: Molecular Insights and Potential Therapeutic Applications. Curr. Neuropharmacol. 2023, 21, 1184–1201. [Google Scholar] [CrossRef]
  145. Pinyaev, S.I.; Kuzmenko, T.P.; Revina, N.V.; Parchaykina, M.V.; Pronin, A.S.; Syusin, I.V.; Novozhilova, O.S.; Revin, V.V.; Chudaikina, E.V.; Revina, E.S. Influence of Resveratrol on Oxidation Processes and Lipid Phase Characteristics in Damaged Somatic Nerves. Biomed. Res. Int. 2019, 2019, 2381907. [Google Scholar] [CrossRef]
  146. Zhao, H.; Wang, Q.; Cheng, X.; Li, X.; Li, N.; Liu, T.; Li, J.; Yang, Q.; Dong, R.; Zhang, Y. Inhibitive Effect of Resveratrol on the Inflammation in Cultured Astrocytes and Microglia Induced by Aβ1–42. Neuroscience 2018, 379, 390–404. [Google Scholar] [CrossRef]
  147. Rahman, M.; Akter, R.; Bhattacharya, T.; Abdel-Daim, M.M.; Alkahtani, S.; Arafah, M.W.; Al-Johani, N.S.; Alhoshani, N.M.; Alkeraishan, N.; Alhenaky, A. Resveratrol and Neuroprotection: Impact and Its Therapeutic Potential in Alzheimer’s Disease. Front. Pharmacol. 2020, 11, 619024. [Google Scholar] [CrossRef] [PubMed]
  148. Lama, A.; Pirozzi, C.; Avagliano, C.; Annunziata, C.; Mollica, M.P.; Calignano, A.; Meli, R.; Mattace Raso, G. Nutraceuticals: An Integrative Approach to Starve Parkinson’s Disease. Brain Behav. Immun. Health 2020, 2, 100037. [Google Scholar] [CrossRef] [PubMed]
  149. Lofrumento, D.D.; Nicolardi, G.; Cianciulli, A.; Nuccio, F.; Pesa, V.; Carofiglio, V.; Dragone, T.; Calvello, R.; Panaro, M.A. Neuroprotective Effects of Resveratrol in an MPTP Mouse Model of Parkinson’s-like Disease: Possible Role of SOCS-1 in Reducing pro-Inflammatory Responses. Innate Immun. 2014, 3, 249–260. [Google Scholar] [CrossRef]
  150. Akbari, M.; Lankarani, K.B.; Tabrizi, R.; Ghayour-Mobarhan, M.; Peymani, P.; Ferns, G.; Ghaderi, A.; Asemi, Z. The Effects of Curcumin on Weight Loss Among Patients With Metabolic Syndrome and Related Disorders: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Front. Pharmacol. 2019, 10, 649. [Google Scholar] [CrossRef]
  151. Soetikno, V.; Sari, F.R.; Veeraveedu, P.T.; Thandavarayan, R.A.; Harima, M.; Sukumaran, V.; Lakshmanan, A.P.; Suzuki, K.; Kawachi, H.; Watanabe, K. Curcumin Ameliorates Macrophage Infiltration by Inhibiting NF-κB Activation and Proinflammatory Cytokines in Streptozotocin Induced-Diabetic Nephropathy. Nutr. Metab. 2011, 8, 35. [Google Scholar] [CrossRef]
  152. Zhao, D.; Pan, Y.; Yu, N.; Bai, Y.; Ma, R.; Mo, F.; Zuo, J.; Chen, B.; Jia, Q.; Zhang, D. Curcumin Improves Adipocytes Browning and Mitochondrial Function in 3T3-L1 Cells and Obese Rodent Model. R. Soc. Open Sci. 2021, 3, 200974. [Google Scholar] [CrossRef]
  153. Motaghinejad, M.; Motevalian, M.; Fatima, S.; Hashemi, H.; Gholami, M. Curcumin Confers Neuroprotection against Alcohol-Induced Hippocampal Neurodegeneration via CREB-BDNF Pathway in Rats. Biomed. Pharmacother. 2017, 87, 721–740. [Google Scholar] [CrossRef] [PubMed]
  154. Eckert, G.P.; Schiborr, C.; Hagl, S.; Abdel-Kader, R.; Müller, W.E.; Rimbach, G.; Frank, J. Curcumin Prevents Mitochondrial Dysfunction in the Brain of the Senescence-Accelerated Mouse-Prone 8. Neurochem. Int. 2013, 62, 595–602. [Google Scholar] [CrossRef]
  155. Kabir, M.; Rahman, M.; Shah, M.; Jamiruddin, M.; Basak, D.; Al-Harrasi, A.; Bhatia, S.; Ashraf, G.M.; Najda, A.; El-kott, A.F. Therapeutic Promise of Carotenoids as Antioxidants and Anti-Inflammatory Agents in Neurodegenerative Disorders. Biomed. Pharmacother. 2022, 146, 112610. [Google Scholar] [CrossRef] [PubMed]
  156. Ademowo, O.S.; Oyebode, O.; Edward, R.; Conway, M.E.; Griffiths, H.R.; Dias, I.H. Effects of Carotenoids on Mitochondrial Dysfunction. Biochem. Soc. Trans. 2024, 1, 65–74. [Google Scholar] [CrossRef]
  157. Canas, J.A.; Lochrie, A.; McGowan, A.G.; Hossain, J.; Schettino, C.; Balagopal, P.B. Effects of Mixed Carotenoids on Adipokines and Abdominal Adiposity in Children: A Pilot Study. J. Clin. Endocrinol. Metab. 2017, 102, 1983–1990. [Google Scholar] [CrossRef] [PubMed]
  158. Kakutani, R.; Hokari, S.; Nishino, A.; Ichihara, T.; Sugimoto, K.; Takaha, T.; Kuriki, T.; Maoka, T. Effect of Oral Paprika Xanthophyll Intake on Abdominal Fat in Healthy Overweight Humans: A Randomized, Double-Blind, Placebo-Controlled Study. J. Oleo Sci. 2018, 9, 1149–1162. [Google Scholar] [CrossRef]
  159. Kawada, T.; Kamei, Y.; Fujita, A.; Hida, Y.; Takahashi, N.; Sugimoto, E.; Fushiki, T. Carotenoids and Retinoids as Suppressors on Adipocyte Differentiation via Nuclear Receptors. Biofactors 2000, 13, 103–109. [Google Scholar] [CrossRef]
  160. Craft, N.E.; Haitema, T.B.; Garnett, K.M.; Fitch, K.A.; Dorey, C.K. Carotenoid, Tocopherol, and Retinol Concentrations in Elderly Human Brain. J. Nutr. Health Aging 2004, 8, 156–162. [Google Scholar]
  161. Takayama, K.; Nishiko, E.; Matsumoto, G.; Inakuma, T. Study on the Expression of C-Fos Protein in the Brain of Rats after Ingestion of Food Rich in Lycopene. Neurosci. Lett. 2013, 536, 1–5. [Google Scholar] [CrossRef]
  162. Lakey-Beitia, J.; Kumar, D.J.; Hegde, M.L.; Rao, K.S. Carotenoids as Novel Therapeutic Molecules Against Neurodegenerative Disorders: Chemistry and Molecular Docking Analysis. Int. J. Mol. Sci. 2019, 20, 5553. [Google Scholar] [CrossRef]
  163. Wu, L.; Lu, P.; Guo, X.; Song, K.; Lyu, Y.; Bothwell, J.; Wu, J.; Hawkins, O.; Clarke, S.L.; Lucas, E.A. β-Carotene Oxygenase 2 Deficiency-Triggered Mitochondrial Oxidative Stress Promotes Low-Grade Inflammation and Metabolic Dysfunction. Free Radic. Biol. Med. 2021, 164, 271–284. [Google Scholar] [CrossRef] [PubMed]
  164. Zhu, R.; Wei, J.; Liu, H.; Liu, C.; Wang, L.; Chen, B.; Li, L.; Jia, Q.; Tian, Y.; Li, R. Lycopene Attenuates Body Weight Gain through Induction of Browning via Regulation of Peroxisome Proliferator-Activated Receptor γ in High-Fat Diet-Induced Obese Mice. J. Nutr. Biochem. 2020, 78, 108335. [Google Scholar] [CrossRef] [PubMed]
  165. Wang, J.; Suo, Y.; Zhang, J.; Zou, Q.; Tan, X.; Yuan, T.; Liu, Z.; Liu, X. Lycopene Supplementation Attenuates Western Diet-Induced Body Weight Gain through Increasing the Expressions of Thermogenic/Mitochondrial Functional Genes and Improving Insulin Resistance in the Adipose Tissue of Obese Mice. J. Nutr. Biochem. 2019, 69, 63–72. [Google Scholar] [CrossRef]
  166. Yu, L.; Wang, W.; Pang, W.; Xiao, Z.; Jiang, Y.; Hong, Y. Dietary Lycopene Supplementation Improves Cognitive Performances in Tau Transgenic Mice Expressing P301L Mutation via Inhibiting Oxidative Stress and Tau Hyperphosphorylation. J. Alzheimers Dis. 2017, 57, 475–482. [Google Scholar] [CrossRef]
  167. Yi, F.; He, X.; Wang, D. Lycopene Protects against MPP(+)-Induced Cytotoxicity by Maintaining Mitochondrial Function in SH-SY5Y Cells. Neurochem. Res. 2013, 8, 1747–1757. [Google Scholar] [CrossRef]
  168. Prakash, A.; Kumar, A. Implicating the Role of Lycopene in Restoration of Mitochondrial Enzymes and BDNF Levels in β-Amyloid Induced Alzheimer׳s Disease. Eur. J. Pharmacol. 2014, 741, 104–111. [Google Scholar] [CrossRef]
  169. Choi, S.A.; Oh, Y.K.; Lee, J.; Sim, S.J.; Hong, M.E.; Park, J.Y.; Kim, M.S.; Kim, S.W.; Lee, J.S. High-Efficiency Cell Disruption and Astaxanthin Recovery from Haematococcus Pluvialis Cyst Cells Using Room-Temperature Imidazolium-Based Ionic Liquid/Water Mixtures. Bioresour Technol 2019, 274, 120–126. [Google Scholar] [CrossRef] [PubMed]
  170. Ikeuchi, M.; Koyama, T.; Takahashi, J.; Yazawa, K. Effects of astaxanthin in obese mice fed a high-fat diet. Biosci. Biotechnol. Biochem. 2007, 71, 893–899. [Google Scholar] [CrossRef]
  171. Nishida, Y.; Nawaz, A.; Kado, T.; Takikawa, A.; Igarashi, Y.; Onogi, Y.; Wada, T.; Sasaoka, T.; Yamamoto, S.; Sasahara, M. Astaxanthin Stimulates Mitochondrial Biogenesis in Insulin Resistant Muscle via Activation of AMPK Pathway. J. Cachexia Sarcopenia Muscle 2020, 1, 241–258. [Google Scholar] [CrossRef]
  172. Kohandel, Z.; Farkhondeh, T.; Aschner, M.; Pourbagher-Shahri, A.M.; Samarghandian, S. Anti-Inflammatory Action of Astaxanthin and Its Use in the Treatment of Various Diseases. Biomed. Pharmacother. 2022, 145, 112179. [Google Scholar] [CrossRef]
  173. Wang, S.; Qi, X. The Putative Role of Astaxanthin in Neuroinflammation Modulation: Mechanisms and Therapeutic Potential. Front. Pharmacol. 2022, 13, 916653. [Google Scholar] [CrossRef]
  174. Kuo, M.H.; Lee, H.F.; Tu, Y.F.; Lin, L.H.; Cheng, Y.Y.; Lee, H.T. Astaxanthin Ameliorates Ischemic-Hypoxic-Induced Neurotrophin Receptor P75 Upregulation in the Endothelial Cells of Neonatal Mouse Brains. Int. J. Mol. Sci. 2019, 24, 6168. [Google Scholar] [CrossRef] [PubMed]
  175. Che, H.; Li, Q.; Zhang, T.; Wang, D.; Yang, L.; Xu, J.; Yanagita, T.; Xue, C.; Chang, Y.; Wang, Y. Effects of Astaxanthin and Docosahexaenoic-Acid-Acylated Astaxanthin on Alzheimer’s Disease in APP/PS1 Double-Transgenic Mice. J. Agric. Food Chem. 2018, 66, 4948–4957. [Google Scholar] [CrossRef] [PubMed]
  176. Custers, E.M.; Kiliaan, A.J. Dietary lipids from body to brain. Prog. Lipid Res. 2022, 85, 101144. [Google Scholar] [CrossRef]
  177. Spagnuolo, M.S.; Maresca, B.; Mollica, M.P.; Cavaliere, G.; Cefaliello, C.; Trinchese, G.; Esposito, M.G.; Scudiero, R.; Crispino, M.; Abrescia, P. Haptoglobin Increases with Age in Rat Hippocampus and Modulates Apolipoprotein E Mediated Cholesterol Trafficking in Neuroblastoma Cell Lines. Front. Cell Neurosci. 2014, 8, 212. [Google Scholar] [CrossRef]
  178. Cavaliere, G.; Trinchese, G.; Bergamo, P.; De Filippo, C.; Mattace Raso, G.; Gifuni, G.; Putti, R.; Moni, B.H.; Canani, R.B.; Meli, R.; et al. Polyunsaturated Fatty Acids Attenuate Diet Induced Obesity and Insulin Resistance, Modulating Mitochondrial Respiratory Uncoupling in Rat Skeletal Muscle. PLoS ONE 2016, 11, e0149033. [Google Scholar] [CrossRef] [PubMed]
  179. Lionetti, L.; Mollica, M.P.; Donizzetti, I.; Gifuni, G.; Sica, R.; Pignalosa, A.; Cavaliere, G.; Gaita, M.; Filippo, C.; Zorzano, A. High-Lard and High-Fish-Oil Diets Differ in Their Effects on Function and Dynamic Behaviour of Rat Hepatic Mitochondria. PLoS ONE 2014, 3, e92753. [Google Scholar] [CrossRef]
  180. Calon, F.; Cole, G. Neuroprotective Action of Omega-3 Polyunsaturated Fatty Acids against Neurodegenerative Diseases: Evidence from Animal Studies. Prostaglandins Leukot. Essent. Fatty Acids 2007, 77, 287–293. [Google Scholar] [CrossRef]
  181. Xiao, M.; Xiang, W.; Chen, Y.; Peng, N.; Du, X.; Lu, S.; Zuo, Y.; Li, B.; Hu, Y.; Li, X. DHA Ameliorates Cognitive Ability, Reduces Amyloid Deposition, and Nerve Fiber Production in Alzheimer’s Disease. Front. Nutr. 2022, 9, 852433. [Google Scholar] [CrossRef]
  182. De Lau, L.M.L.; Bornebroek, M.; Witteman, J.C.M.; Hofman, A.; Koudstaal, P.J.; Breteler, M.M.B. Dietary Fatty Acids and the Risk of Parkinson Disease: The Rotterdam Study. Neurology 2005, 64, 2040–2045. [Google Scholar] [CrossRef]
  183. Mett, J. The Impact of Medium Chain and Polyunsaturated ω-3-Fatty Acids on Amyloid-β Deposition, Oxidative Stress and Metabolic Dysfunction Associated with Alzheimer’s Disease. Antioxidants 2021, 10, 1991. [Google Scholar] [CrossRef]
  184. Li, P.; Song, C. Potential Treatment of Parkinson’s Disease with Omega-3 Polyunsaturated Fatty Acids. Nutr. Neurosci. 2022, 25, 180–191. [Google Scholar] [CrossRef]
  185. Lionetti, L.; Mollica, M.; Sica, R.; Donizzetti, I.; Gifuni, G.; Pignalosa, A.; Cavaliere, G.; Putti, R. Differential Effects of High-Fish Oil and High-Lard Diets on Cells and Cytokines Involved in the Inflammatory Process in Rat Insulin-Sensitive Tissues. Int. J. Mol. Sci. 2014, 15, 3040–3063. [Google Scholar] [CrossRef] [PubMed]
  186. Trinchese, G.; Feola, A.; Cavaliere, G.; Cimmino, F.; Catapano, A.; Penna, E.; Scala, G.; Greco, L.; Bernardo, L.; Porcellini, A.; et al. Mitochondrial Metabolism and Neuroinflammation in the Cerebral Cortex and Cortical Synapses of Rats: Effect of Milk Intake through DNA Methylation. J. Nutr. Biochem. 2024, 128, 109624. [Google Scholar] [CrossRef] [PubMed]
  187. Trinchese, G.; Cavaliere, G.; De Filippo, C.; Aceto, S.; Prisco, M.; Chun, J.T.; Penna, E.; Negri, R.; Muredda, L.; Demurtas, A.; et al. Human Milk and Donkey Milk, Compared to Cow Milk, Reduce Inflammatory Mediators and Modulate Glucose and Lipid Metabolism, Acting on Mitochondrial Function and Oleylethanolamide Levels in Rat Skeletal Muscle. Front. Physiol. 2018, 9, 32. [Google Scholar] [CrossRef] [PubMed]
  188. Trinchese, G.; Cavaliere, G.; Cimmino, F.; Catapano, A.; Carta, G.; Pirozzi, C.; Murru, E.; Lama, A.; Meli, R.; Bergamo, P.; et al. Decreased Metabolic Flexibility in Skeletal Muscle of Rat Fed with a High-Fat Diet Is Recovered by Individual CLA Isomer Supplementation via Converging Protective Mechanisms. Cells 2020, 9, 823. [Google Scholar] [CrossRef]
  189. Mollica, M.P.; Trinchese, G.; Cavaliere, G.; De Filippo, C.; Cocca, E.; Gaita, M.; Della-Gatta, A.; Marano, A.; Mazzarella, G.; Bergamo, P. C9,T11-Conjugated Linoleic Acid Ameliorates Steatosis by Modulating Mitochondrial Uncoupling and Nrf2 Pathway. J. Lipid Res. 2014, 55, 837–849. [Google Scholar] [CrossRef]
  190. Lehnen, T.E.; Silva, M.R.; Camacho, A.; Marcadenti, A.; Lehnen, A.M. A Review on Effects of Conjugated Linoleic Fatty Acid (CLA) upon Body Composition and Energetic Metabolism. J. Int. Soc. Sports Nutr. 2015, 12, 36. [Google Scholar] [CrossRef]
  191. Shen, W.; Baldwin, J.; Collins, B.; Hixson, L.; Lee, K.T.; Herberg, T.; Starnes, J.; Cooney, P.; Chuang, C.C.; Hopkins, R. Low Level of Trans-10, Cis-12 Conjugated Linoleic Acid Decreases Adiposity and Increases Browning Independent of Inflammatory Signaling in Overweight Sv129 Mice. J. Nutr. Biochem. 2015, 6, 616–625. [Google Scholar] [CrossRef]
  192. Aydın, B.; Güler Şahin, C.; Şekeroğlu, V.; Atlı Şekeroğlu, Z. Conjugated Linoleic Acid Protects Brain Mitochondrial Function in Acrolein Induced Male Rats. Toxicol. Mech. Methods 2021, 31, 674–679. [Google Scholar] [CrossRef]
  193. Cavaliere, G.; Trinchese, G.; Musco, N.; Infascelli, F.; Filippo, C.; Mastellone, V.; Morittu, V.M.; Lombardi, P.; Tudisco, R.; Grossi, M. Milk from Cows Fed a Diet with a High Forage:Concentrate Ratio Improves Inflammatory State, Oxidative Stress, and Mitochondrial Function in Rats. J. Dairy. Sci. 2018, 101, 1843–1851. [Google Scholar] [CrossRef] [PubMed]
  194. Cuciniello, R.; Luongo, D.; Ferramosca, A.; Lunetti, P.; Rotondi-Aufiero, V.; Crispi, S.; Zara, V.; Maurano, F.; Filosa, S.; Bergamo, P. Conjugated Linoleic Acid Downregulates Alzheimer’s Hallmarks in Aluminum Mouse Model through an Nrf2-Mediated Adaptive Response and Increases Brain Glucose Transporter Levels. Free Radic. Biol. Med. 2022, 191, 48–58. [Google Scholar] [CrossRef]
  195. Xiong, R.G.; Zhou, D.D.; Wu, S.X.; Huang, S.Y.; Saimaiti, A.; Yang, Z.J.; Shang, A.; Zhao, C.N.; Gan, R.Y.; Li, H.B. Health Benefits and Side Effects of Short-Chain Fatty Acids. Foods 2022, 18, 2863. [Google Scholar] [CrossRef] [PubMed]
  196. Anachad, O.; Taouil, A.; Taha, W.; Bennis, F.; Chegdani, F. The Implication of Short-Chain Fatty Acids in Obesity and Diabetes. Microbiol. Insights 2023, 16, 11786361231162720. [Google Scholar] [CrossRef] [PubMed]
  197. Mollica, M.P.; Mattace Raso, G.; Cavaliere, G.; Trinchese, G.; De Filippo, C.; Aceto, S.; Prisco, M.; Pirozzi, C.; Di Guida, F.; Lama, A.; et al. Butyrate Regulates Liver Mitochondrial Function, Efficiency, and Dynamics in Insulin-Resistant Obese Mice. Diabetes 2017, 66, 1405–1418. [Google Scholar] [CrossRef]
  198. Chakraborty, P.; Gamage, H.K.A.H.; Laird, A.S. Butyrate as a Potential Therapeutic Agent for Neurodegenerative Disorders. Neurochem. Int. 2024, 176, 105745. [Google Scholar] [CrossRef]
  199. Avagliano, C.; Coretti, L.; Lama, A.; Pirozzi, C.; Caro, C.; Biase, D.; Turco, L.; Mollica, M.P.; Paciello, O.; Calignano, A. Dual-Hit Model of Parkinson’s Disease: Impact of Dysbiosis on 6-Hydroxydopamine-Insulted Mice—Neuroprotective and Anti-Inflammatory Effects of Butyrate. Int. J. Mol. Sci. 2022, 23, 6367. [Google Scholar] [CrossRef]
  200. Pirozzi, C.; Lama, A.; Annunziata, C.; Cavaliere, G.; Caro, C.; Citraro, R.; Russo, E.; Tallarico, M.; Iannone, M.; Ferrante, M.C. Butyrate Prevents Valproate-induced Liver Injury: In Vitro and in Vivo Evidence. FASEB J. 2020, 34, 676–690. [Google Scholar] [CrossRef]
  201. Jiang, Y.; Li, K.; Li, X.; Xu, L.; Yang, Z. Sodium Butyrate Ameliorates the Impairment of Synaptic Plasticity by Inhibiting the Neuroinflammation in 5XFAD Mice. Chem. Biol. Interact. 2021, 341, 109452. [Google Scholar] [CrossRef]
  202. Thabuis, C.; Destaillats, F.; Tissot-Favre, D.; Martin, J.C. Oleoyl-ethanolamide (OEA): A bioactive lipid derived from oleic acid and phosphatidylethanol-amine. Lipid Technol. 2007, 19, 225–227. [Google Scholar] [CrossRef]
  203. Beggiato, S.; Tomasini, M.C.; Ferraro, L. Palmitoylethanolamide (PEA) as a Potential Therapeutic Agent in Alzheimer’s Disease. Front. Pharmacol. 2019, 10, 821. [Google Scholar] [CrossRef] [PubMed]
  204. Mattace Raso, G.; Santoro, A.; Russo, R.; Simeoli, R.; Paciello, O.; Carlo, C.; Diano, S.; Calignano, A.; Meli, R. Palmitoylethanolamide Prevents Metabolic Alterations and Restores Leptin Sensitivity in Ovariectomized Rats. Endocrinology 2014, 4, 1291–1301. [Google Scholar] [CrossRef]
  205. Annunziata, C.; Pirozzi, C.; Lama, A.; Senzacqua, M.; Comella, F.; Bordin, A.; Monnolo, A.; Pelagalli, A.; Ferrante, M.C.; Mollica, M.P. Palmitoylethanolamide Promotes White-to-Beige Conversion and Metabolic Reprogramming of Adipocytes: Contribution of PPAR-α. Pharmaceutics 2022, 14, 338. [Google Scholar] [CrossRef] [PubMed]
  206. Lama, A.; Pirozzi, C.; Severi, I.; Morgese, M.G.; Senzacqua, M.; Annunziata, C.; Comella, F.; Del Piano, F.; Schiavone, S.; Petrosino, S. Palmitoylethanolamide Dampens Neuroinflammation and Anxiety-like Behavior in Obese Mice. Brain Behav. Immun. 2022, 102, 110–123. [Google Scholar] [CrossRef]
  207. Cristiano, C.; Pirozzi, C.; Coretti, L.; Cavaliere, G.; Lama, A.; Russo, R.; Lembo, F.; Mollica, M.P.; Meli, R.; Calignano, A. Palmitoylethanolamide Counteracts Autistic-like Behaviours in BTBR T+tf/J Mice: Contribution of Central and Peripheral Mechanisms. Brain Behav. Immun. 2018, 74, 166–175. [Google Scholar] [CrossRef]
  208. Annunziata, C.; Lama, A.; Pirozzi, C.; Cavaliere, G.; Trinchese, G.; Guida, F.; Nitrato Izzo, A.; Cimmino, F.; Paciello, O.; Biase, D. Palmitoylethanolamide Counteracts Hepatic Metabolic Inflexibility Modulating Mitochondrial Function and Efficiency in Diet-induced Obese Mice. FASEB J. 2020, 34, 350–364. [Google Scholar] [CrossRef]
  209. Laleh, P.; Yaser, K.; Abolfazl, B.; Shahriar, A.; Mohammad, A.J.; Nazila, F.; Alireza, O. Oleoylethanolamide Increases the Expression of PPAR-A and Reduces Appetite and Body Weight in Obese People: A Clinical Trial. Appetite 2018, 128, 44–49. [Google Scholar] [CrossRef] [PubMed]
  210. Comerota, M.M.; Gedam, M.; Xiong, W.; Jin, F.; Deng, L.; Wang, M.C.; Wang, J.; Zheng, H. Oleoylethanolamide Facilitates PPARα and TFEB Signaling and Attenuates Aβ Pathology in a Mouse Model of Alzheimer’s Disease. Mol. Neurodegener. 2023, 18, 56. [Google Scholar] [CrossRef]
  211. Gonzalez-Aparicio, R.; Blanco, E.; Serrano, A.; Pavon, F.J.; Parsons, L.H.; Maldonado, R.; Robledo, P.; Fernandez-Espejo, E.; Fonseca, F.R. The Systemic Administration of Oleoylethanolamide Exerts Neuroprotection of the Nigrostriatal System in Experimental Parkinsonism. Int. J. Neuropsychopharmacol. 2014, 3, 455–468. [Google Scholar] [CrossRef]
  212. Reyes-Soto, C.Y.; Villaseca-Flores, M.; Ovalle-Noguez, E.A.; Nava-Osorio, J.; Galván-Arzate, S.; Rangel-López, E.; Maya-López, M.; Retana-Márquez, S.; Túnez, I.; Tinkov, A.A. Oleamide Reduces Mitochondrial Dysfunction and Toxicity in Rat Cortical Slices Through the Combined Action of Cannabinoid Receptors Activation and Induction of Antioxidant Activity. Neurotox. Res. 2022, 40, 2167–2178. [Google Scholar] [CrossRef]
  213. Errazzino, S.; Berto, F.; Dalle Carbonare, M.; Fabris, M.; Guiotto, A.; Bernardini, D.; Leon, A. Stearoylethanolamide Exerts Anorexic Effects in Mice via Down-Regulation of Liver Stearoyl-Coenzyme A Desaturase-1 mRNA Expression. FASEB J. 2004, 13, 1580–1582. [Google Scholar] [CrossRef] [PubMed]
  214. Tkachenko, O.; Kosiakova, H.; Klimashevsky, V.; Berdyshev, A.; Hula, N. The Effect of N-Stearoylethanolamine on the Adipocyte Fatty Acid Composition of Different Age Rats with Obesity-Induced Insulin Resistance. EUREKA Life Sci. 2020, 2, 10–23. [Google Scholar] [CrossRef]
  215. Lykhmus, O.; Uspenska, K.; Koval, L.; Lytovchenko, D.; Voytenko, L.; Horid’ko, T.; Kosiakova, H.; Gula, N.; Komisarenko, S.; Skok, M. N-Stearoylethanolamine Protects the Brain and Improves Memory of Mice Treated with Lipopolysaccharide or Immunized with the Extracellular Domain of A7 Nicotinic Acetylcholine Receptor. Int. Immunopharmacol. 2017, 52, 290–296. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Link between mitochondrial dysfunction in adipose tissue and progression of neurodegenerative diseases. In hypertrophic adipocytes mitochondrial dysfunction leads to increased ROS production, resulting in increased release of pro-inflammatory cytokines, such as tumor necrosis factor alpha (TNF-α) and interleukin-6 (IL-6). The inflammatory state is transmitted via the systemic circulation to the central nervous system (CNS), where it impairs neuronal mitochondrial function and leads to the development of neurodegenerative diseases (NDDs). Alzheimer’s disease (AD), Parkinson’s disease (PD), multiple sclerosis (MS).
Figure 1. Link between mitochondrial dysfunction in adipose tissue and progression of neurodegenerative diseases. In hypertrophic adipocytes mitochondrial dysfunction leads to increased ROS production, resulting in increased release of pro-inflammatory cytokines, such as tumor necrosis factor alpha (TNF-α) and interleukin-6 (IL-6). The inflammatory state is transmitted via the systemic circulation to the central nervous system (CNS), where it impairs neuronal mitochondrial function and leads to the development of neurodegenerative diseases (NDDs). Alzheimer’s disease (AD), Parkinson’s disease (PD), multiple sclerosis (MS).
Biomolecules 15 00638 g001
Figure 2. Role of biomolecules in preventing or reversing obesity and neurodegenerative disorders. Polyphenols, carotenoids, ω-3 polyunsaturated fatty acids (ω-3 PUFAs), short-chain fatty acids (SCFAs), and endocannabinoid-like compounds (oleoylethanolamide (OEA), palmitoylethanolamide (PEA), and stearoylethanolamide (SEA)) prevent or reverse obesity and neurodegenerative disorders (NDDs) by counteracting adipogenesis, ROS production, and neuronal death and by increasing mitochondrial function.
Figure 2. Role of biomolecules in preventing or reversing obesity and neurodegenerative disorders. Polyphenols, carotenoids, ω-3 polyunsaturated fatty acids (ω-3 PUFAs), short-chain fatty acids (SCFAs), and endocannabinoid-like compounds (oleoylethanolamide (OEA), palmitoylethanolamide (PEA), and stearoylethanolamide (SEA)) prevent or reverse obesity and neurodegenerative disorders (NDDs) by counteracting adipogenesis, ROS production, and neuronal death and by increasing mitochondrial function.
Biomolecules 15 00638 g002
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

Marino, F.; Petrella, L.; Cimmino, F.; Pizzella, A.; Monda, A.; Allocca, S.; Rotondo, R.; D’Angelo, M.; Musco, N.; Iommelli, P.; et al. From Obesity to Mitochondrial Dysfunction in Peripheral Tissues and in the Central Nervous System. Biomolecules 2025, 15, 638. https://doi.org/10.3390/biom15050638

AMA Style

Marino F, Petrella L, Cimmino F, Pizzella A, Monda A, Allocca S, Rotondo R, D’Angelo M, Musco N, Iommelli P, et al. From Obesity to Mitochondrial Dysfunction in Peripheral Tissues and in the Central Nervous System. Biomolecules. 2025; 15(5):638. https://doi.org/10.3390/biom15050638

Chicago/Turabian Style

Marino, Francesca, Lidia Petrella, Fabiano Cimmino, Amelia Pizzella, Antonietta Monda, Salvatore Allocca, Roberta Rotondo, Margherita D’Angelo, Nadia Musco, Piera Iommelli, and et al. 2025. "From Obesity to Mitochondrial Dysfunction in Peripheral Tissues and in the Central Nervous System" Biomolecules 15, no. 5: 638. https://doi.org/10.3390/biom15050638

APA Style

Marino, F., Petrella, L., Cimmino, F., Pizzella, A., Monda, A., Allocca, S., Rotondo, R., D’Angelo, M., Musco, N., Iommelli, P., Catapano, A., Bagnato, C., Paolini, B., & Cavaliere, G. (2025). From Obesity to Mitochondrial Dysfunction in Peripheral Tissues and in the Central Nervous System. Biomolecules, 15(5), 638. https://doi.org/10.3390/biom15050638

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop